tesis doctoral - Dadun - Universidad de Navarra [PDF]

UNIVERSIDAD DE NAVARRA. TESIS DOCTORAL. “Nanosystems for the oral administration of alkyl-‐ lysophospholipid anti-‐tumor

0 downloads 6 Views 11MB Size

Recommend Stories


Universidad de Navarra - Dadun
In the end only three things matter: how much you loved, how gently you lived, and how gracefully you

universidad de granada tesis doctoral
The best time to plant a tree was 20 years ago. The second best time is now. Chinese Proverb

Universidad de Huelva Tesis doctoral
Learning never exhausts the mind. Leonardo da Vinci

universidad de córdoba tesis doctoral
Suffering is a gift. In it is hidden mercy. Rumi

universidad de córdoba tesis doctoral
You often feel tired, not because you've done too much, but because you've done too little of what sparks

tesis doct tesis doctoral - Universidad Autónoma de Madrid [PDF]
Levstik, 2008; Carretero, 2011; Hobsbawm, 1997; Wertsch, 2002). En este sentido, la enseñanza de la ...... de que se trata de tradiciones inventadas, en la línea que proponen Hobsbawm y Ranger. (1983), cuyo objetivo es legitimar a ...... Las revolu

Universidad Jaume I de Castellón Tesis Doctoral
Life is not meant to be easy, my child; but take courage: it can be delightful. George Bernard Shaw

universidad complutense de madrid tesis doctoral
Live as if you were to die tomorrow. Learn as if you were to live forever. Mahatma Gandhi

tesis doctoral 2015 universidad de granada
Courage doesn't always roar. Sometimes courage is the quiet voice at the end of the day saying, "I will

tesis doctoral 2015 universidad de granada
So many books, so little time. Frank Zappa

Idea Transcript


Departamento  de  Farmacia  y  Tecnología  Farmacéutica   Facultad  de  Farmacia  

 DE  NAVARRA    

 

TESIS  DOCTORAL   “Nanosystems  for  the  oral  administration  of  alkyl-­‐ lysophospholipid  anti-­‐tumor  agents:  development   of  lipid  nanotransporters  and  preclinical  studies”    

 

 

Trabajo  presentado  por  Ander  Estella  Hermoso  de  Mendoza  para   obtener  el  Grado  de  Doctor                              

 

Fdo.  Ander  Estella  Hermoso  de  Mendoza   Pamplona,  2011  

Servicio de Publicaciones de la Universidad de Navarra ISBN 84-8081-097-1

Dña.  Mª  José  Blanco  Prieto,  Profesora  Investigadora  de  la  Universidad  de  Navarra,   informa  que:   El  presente  trabajo:  “Nanosystems  for  the  oral  administration  of  alkyl-­‐ lysophospholipid  

anti-­‐tumor  

agents:  

development  

of  

lipid  

nanotransporters   and   preclinical   studies”   presentado   por   D.   Ander   Estella   Hermoso   de   Mendoza   para   optar   al   grado   de   Doctor,   ha   sido   realizado   bajo   su   dirección   en   el   Departamento   de   Farmacia   y   Tecnología  Farmacéutica  de  la  Facultad  de  Farmacia  de  la  Universidad   de  Navarra  y,  una  vez  revisado,  no  encuentra  objeciones  para  que  sea   presentado  a  su  lectura  y  defensa.   Y  para  que  así  conste,  firma  el  presente  informe.           Fdo.  Dra.  María  José  Blanco  Prieto   Pamplona,  2011          

 

                        Las   investigaciones   realizadas   en   el   presente   trabajo   se   han   llevado   a   cabo   dentro   de   los   proyectos   del   Ministerio   de   Ciencia   e   Innovación   (SAF2007-­‐61261,   SAF2010-­‐15547,   PCT-­‐ 090100-­‐2007-­‐27)   del   Gobierno   de   España,   de   la   Fundación   Caja   Navarra,   Ibercaja,   del   Departamento   de   Salud   del   Gobierno  de  Navarra  (beca  Ortiz  de  Landázuri,  ref:  63/09)  y  de   la   Línea   Especial   “Nanotecnologías   y   liberación   controlada   de   fármacos”  de  la  Universidad  de  Navarra.     Así   mismo,   agradezco   al   Departamento   de   Educación   del   Gobierno  Vasco  (BFI06.37),  al  Departamento  de  Educación  del   Gobierno   de   Navarra   y   a   la   Asociación   de   Amigos   de   la   Universidad   de   Navarra   las   becas   predoctorales   concedidas   para  la  consecución  de  este  trabajo.    

   

                                        A  mis  padres,  Javier  y  Mª  Jose        

    Muchas  gracias…   A   la   Universidad   de   Navarra   y   al   Departamento   de   Farmacia   y   Tecnología   Farmacéutica,   por   permitirme   realizar   esta   tesis   doctoral.   Al   Gobierno   Vasco   (Dpto.   Educación),   al   Ministerio   de   Ciencia   e   Innovación,   al   Gobierno   de   Navarra,   a   Caja   Navarra   y   a   la   Asociación   de   Amigos   de   la   Universidad   por   la   ayuda  económica  aportada  durante  estos  años.   A  la  Dra.  María  Blanco,  por  guiarme  todos  estos  años,  por  confiar  en  mi  persona  y   por   toda   la   ayuda   que   me   has   ofrecido   estos   años.   Gracias   porque   además   de   directora,  has  sido  amiga  y  me  has  dado  infinidad  de  consejos.  Creo  que  no  podía   haber  caido  en  mejores  manos  para  hacer  esta  tesis.  Muchas  gracias!   A  Faustino  Mollinedo,  por  colaborar  activamente  en  el  diseño  de  experimentos  y   ayudarnos   a   interpretar   los   resultados;   y   a   su   laboratorio   en   el   Centro   de   Investigación  del  Cáncer  de  Salamanca,  por  ayudarme  durante  todas  las  estancias   que   he   realizado   allí,   especialmente   Janis   de   la   Iglesia-­‐Vicente,   porque   sin   su   ayuda   no   habría   podido   realizar   varios   experimentos   de   este   trabajo.   Muchas   gracias,  chati.   Al   Dr.   Miguel   Ángel   Campanero   (Campi),   por   toda   la   ayuda   prestada   durante   todos   estos   años,   que   ha   sido   mucha,   y   por   todos   los   buenísimos   momentos   que   hemos  pasado  en  el  departamento  y  fuera  de  él.  La  cromatografía  no  será  lo  mismo   sin  tu  ayuda…   Al   resto   de   doctores,   profesores   y   personal   del   departamento:   Dña.   Marije   Renedo,   Dña.   Pilar   Ygartua,   Dña.   Carmen   Dios,   Dña.   María   del   Mar   Goñi,   D.   Felix  Recarte,  Dña.  Pilar  Guillén  (por  recordarme  cuántas  pastas  o  croissants  me   comía   al   cabo   de   la   mañana   los   días   que   había   cumpleaños…   un   besazo,   Pili!),   D.   Juan   Luis   Martín,   Dña.   Paula   Oteiza,   D.   Fernando   Martínez,   Dña.   Socorro   Espuelas,   Dña.   Maribel   Calvo,   Dña.   Conchita   Tros,   Dña.   María   Jesús   Garrido,   D.  Iñaki  Fdez.  De  Troconiz  y  D.  Juan  Manuel  Irache.     Dentro   de   este   grupo   tan   amplio,   me   gustaría   dar   las   gracias   especialmente   a   la   Dra.   Pilar   Ygartua,   por   ser   una   gran   persona,   porque   me   ha   escuchado   y   ha   sabido   darme   consejos   durante   toda   mi   andadura,   especialmente   en   este   último   tramo,   porque   personas   como   ella   quedan   pocas…   Muchas   gracias   por   darme   tu   apoyo,  Pilar!  

    Muchas  gracias…   A   mis   compañeros   de   grupo   de   trabajo,   pilares   sobre   los   que   me   he   podido   apoyar   durante  mis  momentos  de  flaqueo,  que  bien  saben  que  han  sido  muchos:     A   Edurne   Imbuluzqueta,   la   mujer   que   más   nombres   ha   tenido   en   éste   nuestro   departamento,   Imbuluz,   Aduren,   Imbulurguete,   Edrine   Imbuluzquetz,…   gracias   de   verdad   por   estar   siempre   ahí,   en   los   buenos   y   malos   momentos,   por   todos   tus   consejos,  por  los  mimos  que  me  has  dado  y  por  enseñarme  que  siempre  se  puede   sacar  algo  bueno  de  las  cosas.  Mila  esker!   A  David  González,  por  tu  compañerismo  sin  límite,  por  dar  todo  por  los  demás  sin   esperar   nada   a   cambio,   por   esos   momentos   de   confidencias   en   el   animalario   y   largas   charlas   que   hemos   tenido   en   el   masas   y   por   todo   el   apoyo   que   me   has   dado.   Un  abrazo,  bordini!     A   Fabio   Rocha,   por   ser   auténtico   (“shi”),   buen   amigo,   buen   compañero,   por   los   ánimos  que  me  has  dado  en  los  momentos  bajos  y  por  los  buenos  momentos  que   hemos  compartido.  Mucho  ánimo,  que  no  te  queda  nada!   A  Eduardo  Ansorena,  que  aunque  ya  no  está  en  el  departamento  (ni  en  el  país)  ha   sido  una  persona  muy  importante  para  mí,  y  ha  formado  parte  junto  con  David  y   conmigo  de  los  míticos  “Bordini”,  gracias  Edu  por  las  risas  que  nos  hemos  echado   juntos  en  tantas  ocasiones  (que  han  sido  muchas  y  en  muchos  lugares  del  mundo!)   y  por  los  ánimos  incondicionales  que  me  has  dado  en  los  momentos  complicadetes.   A   Elisa   Garbayo,   gracias   por   poner   siempre   ese   punto   de   alegría   en   todo   lo   que   hacemos,   por   esa   risa   contagiosa   que   tantas   veces   ha   conseguido   animarme,   por   todas   las   aventuras   que   hemos   tenido   en   estos   años,   porque   aunque   nos   dejaste   una   temporadita,   es   como   si   jamás   te   hubieras   ido.   Te   he   echado   mucho   de   menos,   Elisica!   A  Hugo  Lana,  porque  te  considero  una  parte  fundamental  en  nuestro  grupo  y  en   mi  tesis,  porque  sin  tu  ayuda  esta  tesis  se  habría  quedado  a  la  mitad;  gracias  por   los   infinitos   ratos   que   hemos   pasado   en   el   animalario,   machacando   órganos   y   procesando   muestras,   gracias   por   estar   siempre   ahí,   por   sacar   tiempo   para   tomarte   un   redbull   y   escucharme,   por   ser   tan   descerebrado   como   yo   y   seguirme   el   juego  en  todas  las  “gansadas”  que  nos  hemos  pegado.  GRACIAS,  Lana!!  

 

    Muchas  gracias…   A   los   que   recientemente   habéis   llegado:   Bea   Lasa,   “Blasa”,   que   continuará   mis   pasos  y  por  su  aportación  sanguínea  a  la  ciencia,  Teresa  Simón,  “Maritere”,  por  su   templanza,  Esther  Tamayo,  por  su  inagotable  capacidad  de  trabajo  y  organización   de   reuniones,   y   a   las   últimas   incorporaciones   al   clan,   Cristina   (…)   Tabar   e   Izaskun  Imbuluzqueta  (Imbuluz  Senior!),  mis  competidoras  por  las  meriendas  de   Martín  y  demás  historias.     A   Noelia   Ruz,   porque   empecé   contigo   toda   mi   andadura   en   la   investigación,   gracias  por  enseñarme  todo  lo  que  debía  saber  para  sobrevivir  en  un  laboratorio;   gracias   también   por   todos   los   buenos   ratos   que   hemos   pasado   (casas   rurales,   cenas,…),  por  ser  siempre  tan  buenísima  persona  y  por  tus  consejos.   A   Onintza   Sayar,   muchísimas   gracias   por   todo   tu   apoyo   en   los   8   años   que   he   pasado   en   este   departamento,   porque   SIEMPRE   has   estado   cuando   lo   he   necesitado.   Me   has   ayudado   a   entender   muchas   cosas   de   la   vida.   Gracias   por   enseñarme  que  el  tiempo  lo  cura  todo,  por  los  consejos  (que  han  sido  muchos)  que   me  has  dado  y  por  los  que  todavía  me  vas  a  dar!  Mila  esker,  Onin,  mi  paso  por  el   departamento  no  habría  sido  el  mismo  sin  tu  ayuda.   A  Daniel  Moreno,  Victor  López  y  Cristina  Ederra,  sois  únicos,  lo  sabéis.  Aunque   ya   no   estéis   en   este   departamento,   sabéis   que   formáis   parte   de   él   y   de   mi   tesis.   Gracias   por   ayudarme   en   mis   quebraderos   mentales   y   no   mentales,   por   vuestros   sabios  y  “honorables”  consejos,  por  dar  ese  toque  de  humor  en  los  momentos  más   apropiados.  Os  quiero,  chicos!   A   Raquel   Martins,   mi   “Rachel”,   lo   más   parecido   a   una   hermana   que   he   tenido,   comenzamos   juntos   este   camino,   sufrimos   juntos,   “sobrevivimos”   juntos   y   lo   vamos   a   terminar   juntos…   Muchas   gracias   por   tu   amistad,   tu   comprensión,   tu   compañerismo,  por  todos  los  ratos  que  hemos  disfrutado…  Let’s  dance  now!   A   Cristina   Aranda,   Koldo   Urbiola   y   Lorena   de   Pablo,   voy   a   echar   de   menos   trabajar   con   vosotros,   pero   sobre   todo   las   risas,   las   lecturas   matinales   de   horóscopo,  los  cafés,  en  fin,  todas  aquellas  cosas  que  nos  hacen  inseparables  en  el   tiempo…  como  el  pensar  que  una  tesis  pueden  defenderla  dos  personas…  (Lorena   pitrus  dixit).  

    Muchas  gracias…   A  Patricia  Calleja,  Sara  Zalba,  Judit  Huarte    (la  mac-­‐nífica)  y  Patricia  Ojer,  mis   cuatro   “jinetas”   del   apocalipsis   departamental…   mil   gracias   por   ser   tan   buenas   amigas,  por  todos  los  momentos  de  complicidad  que  hemos  tenido  y  por  todas  las   muestras   de   cariño   que   me   habéis   dado   estos   años.   Gracias   por   todo   vuestro   apoyo,  sabéis  que  me  habéis  ayudado  mucho!     A  la  mac-­‐zona,  Luisa  Ruiz,  Maite  Agüeros  e  Irene  Esparza,  nunca  escribir  en  el   departamento  había  sido  tan  divertido,  gracias  por  vuestro  apoyo  y  consejos,  por   ayudarme   sin   esperar   nada   a   cambio   en   todo   momento,   especialmente   en   los   últimos  días  de  mi  tesis  y  por  todos  los  buenos  ratos.     A   Arianna   Madrid,   María   Matoses,   Zinnia   Parra,   Nieves   Vélez,   Marta   Rayo,   Guiomar   Perez,   Verónica   Madrid,   Sheyla   Rehecho,   Hesham   Salman,   Nacho   Melgar,   Izaskun   Goñi,   Amaya   Lasarte…   y   muchos   más   que   os   podéis   dar   por   agradecidos!     To  Dr.  Véronique  Préat,  and  all  my  colleagues  during  my  stay  in  the  “Unité  de   Pharmacie  Galenique”  of  the  Université  Catholique  de  Louvain  in  Brussels.  Thanks   for  making  me  feel  like  home.   A   Javi   Alonso,   por   aguantarme   todos   estos   años,   gracias   por   tener   siempre   respuesta   a   todas   mis   preguntas,   por   tenderme   tu   mano   en   los   momentos   duros,   que  bien  sabemos  han  sido  muchos.  Quiero  que  sepas  que,  a  pesar  de  todo  lo  que   pase,  buena  parte  de  esta  tesis  te  pertenece.  Gracias  de  corazón.     Y  por  último,  pero  con  diferencia  los  más  importantes  para  mí,  muchísimas  gracias   a   Javier   y   Mª   Jose,   mis   padres,   los   que   siempre   habéis   estado   y   estaréis.   Sé   que,   como   yo,   habéis   pasado   buenos   y   malos   momentos   a   lo   largo   de   esta   tesis,   pero   tal   y  como  me  habéis  enseñado  siempre,  las  cosas  si  se  hacen  despacio  y  con  cabeza   acaban   saliendo   bien…   y   creo   que   este   trabajo   es   muestra   de   ello.   En   serio,   sin   vuestro   apoyo   esta   tesis   no   habría   tenido   sentido   y   por   eso,   os   agradezco   la   fe   que   habéis   tenido   en   mi,   aunque   en   ocasiones   os   haya   parecido   que   iba   sin   rumbo…   Gracias,  de  verdad.    

Y  a  todos  los  que  habéis  aportado  luz  a  mi  camino,  ¡¡gracias  mil!!    

   

                                              "Lo  que  con  mucho  trabajo  se  adquiere,  más  se  ama."  (Aristóteles)  

 

   

 

  Index    

INDEX  .................................................................................................................................................  i   ABBREVIATIONS  /  ABREVIATURAS  ..............................................................................  iii     INTRODUCTION  .........................................................................................................................  1   “Lipid  nanomedicines  for  anticancer  drug  therapy”     HYPOTHESIS  AND  OBJECTIVES  /  HIPOTESIS  Y  OBJETIVOS  ...........................  25     CHAPTER  1  ................................................................................................................................  35   “Antitumor   alkyl   ether   lipid   edelfosine:   tissue   distribution   and   pharmacokinetic   behavior   in   healthy   and   tumor-­‐bearing   immunosuppressed  mice”     CHAPTER  2  .................................................................................................................................  57   “Lipid   nanoparticles   for   alkyl   lysophospholipid   edelfosine   encapsulation:  development  and  in  vitro  characterization”     CHAPTER  3  .................................................................................................................................  81   “Lipid   nanoparticles   loaded   with   alkyl-­‐lysophospholipid   edelfosine:  Pharmacokinetic  profile,  biodistribution  studies  and  in   vivo  efficacy  against  mantle  cell  lymphoma”     CHAPTER  4  ...............................................................................................................  113   “In   vitro   and   in   vivo   efficacy   of   edelfosine-­‐loaded   lipid   nanoparticles  against  glioma”     GENERAL  DISCUSSION  ..........................................................................................  135   DISCUSION  GENERAL  .............................................................................................  175     GENERAL  CONCLUSIONS  ......................................................................................  217   CONCLUSIONES  GENERALES  ...............................................................................  221   FUTURE  PERSPECTIVES  /  PERSPECTIVAS  FUTURAS  ..................................  225  

i  

  Index    

ANNEXES  ....................................................................................................................  229     ANNEX  I  ..........................................................................................................................  231   “Lipid  nanoparticles  in  biomedicine”     ANNEX  II  .........................................................................................................................  257   “Comparative  study  of  a  HPLC–MS  assay  versus  an  UHPLC–MS/MS   for   anti-­‐tumoral   alkyl   lysophospholipid   edelfosine   determination   in  both  biological  samples  and  in  lipid  nanoparticulate  systems”     ANNEX  III  .......................................................................................................................  267   “In   vitro   and   in   vivo   selective   antitumor   activity   of   edelfosine   against   mantle   cell   lymphoma   and   chronic   lymphocytic   leukemia   involving  lipid  rafts”     ANNEX  IV  ........................................................................................................................  279   “Lipid  raft-­‐targeted  therapy  in  multiple  myeloma”     ANNEX  V  .....................................................................................................................  291   Commentary:   “The   Rafts   of   the   Medusa:   cholesterol   targeting   in   cancer  therapy”  

ii  

  Abbreviations  /  Abreviaturas    

AFM  

Atomic  force  microscopy  

Microscopía  de  fuerza  atómica  

AIDS  

Acquired  immune  deficiency  syndrome   Síndrome  de  inmunodeficiencia  adquirida  

ALP  

Alkyl  lysophospholipid  

Alquil  lisofosfolípido  

ApoE  

Apolipoprotein  E  

Apolipoproteína  E  

AUC  

Area  under  the  curve  

Área  bajo  la  curva  

BBB  

Blood  brain  barrier  

Barrera  hematoencefálica  

BSA  

Bovine  serum  albumin  

Albúmina  sérica  bovina  

bw  

Body  weight  

Peso  

CHOL  

Cholesterol  

Colesterol  

CL  

Clearance  

Aclaramiento  

CLL  

Chronic  lymphocytic  leukemia  

Leucemia  linfocítica  crónica  

Cmax  

Peak  concentration  

Concentración  máxima  

Cmin  

Minimum  concentration  

Concentración  mínima  

CSF  

Cerebrospinal  fluid  

Fluido  cerebroespinal  

D  

Dose  

Dosis  

DCP  

Dicetyl  phosphate  

Dicetil  fosfato  

DDAB  

Didecyldimethylammonium  bromide  

Bromuro  de  didecildimetilamonio  

DOPE  

Dioleoylphosphatidyl  ethanolamine  

Dioleoilfosfatidil  etanolamina  

DOTAP  

Dioleoyl  trimethylammonium  propane  

Dioleoil  trimetilamonio  propano  

DSC  

Differential  scanning  calorimetry  

Calorimetría  diferencial  de  barrido  

EDTA  

Ethylenediaminetetraacetic  acid  

Ácido  etilendiaminotetraacético  

ELL-­‐12  

Liposomal  edelfosine  

Edelfosina  liposomal  

EPR  

Enhanced  permeability  and  retention  

Incremento  de  la  permeabilidad  y  retención  

ESI  

Electrospray  ionization  

Ionización  por  electrospray  

ET-­‐18-­‐OCH3   Edelfosine  

Edelfosina  

FBS  

Fetal  bovine  serum  

Suero  fetal  bovino  

FCA  

Freud's  complete  adjuvant  

Adyuvante  completo  de  Freud  

FCS  

Fetal  calf  serum  

Suero  fetal  bovino  

FITC  

Fluorescein  isothiocyanate  

Isotiocianato  de  fluoresceína  

GRAS  

Generally  recognised  as  safe  

Comunmente  reconocidos  como  seguros  

HBV  

Hepatitis  B  virus  

Virus  de  hepatitis  B  

HIV  

Human  immunodeficiency  virus  

Virus  de  inmunodeficiencia  humana  

iii  

  Abbreviations  /  Abreviaturas    

HPLC-­‐MS  

High  performance  liquid   chromatography  mass  spectrometry  

Cromatografía  líquida  de  alta  resolución   acoplada  a  espectrometría  de  masas  

HPTLC  

High  performance  thin  layer   chromatography  

Cromatografía  en  capa  fina  de  alta  resolución  

IARC  

International  Agency  for  Research  on   Cancer  

Agencia  Internacional  de  Investigación  del   Cáncer    

IC50  

Inhibitory  concentration  50  

Concentración  inhibitoria  50  

IFN  

Interferon  

Interferón  

IgG  

Immunoglobulin  G  

Inmunoglobulina  G  

k  

Distribution  constant  rate  

Constante  de  distribución  

LD  

Laser  diffractometry  

Difractometría  láser  

LDC  

Lipid-­‐drug  conjugates  

Conjugados  lípido-­‐fármaco  

LDL  

Low  density  lipoproteins  

Lipoproteinas  de  baja  densidad  

LHRH  

Luteinizing-­‐hormone-­‐  releasing   hormone  

Hormona  liberadora  de  hormona  luteinizante  

LN  

Lipid  nanoparticles  

Nanopartículas  lipídicas  

LOD  

Limit  of  detection  

Límite  de  detección  

LOQ  

Limit  of  quantitation  

Límite  de  cuantificación  

MCD  

Methyl  cyclodextrin  

Metil  ciclodextrina  

MCL  

Mantle  cell  lymphoma  

Linfoma  de  manto  

MDR  

Multidrug  resistant  

Multirresistente  a  fármacos  

MDT  

Mean  dissolution  time  

Tiempo  medio  de  disolución  

MM  

Multiple  myeloma  

Mieloma  múltiple  

MPS  

Mononuclear  phagocytic  system    

Sistema  fagocítico  mononuclear  

MRM  

Multiple  reaction  monitoring  

Monitorización  de  reacciones  múltiples  

MRT  

Mean  residence  time  

Tiempo  medio  de  residencia  

MTT  

3-­‐(4,5-­‐Dimethylthiazol-­‐2-­‐yl)-­‐2,5-­‐ diphenyltetrazolium  bromide  

Bromuro  de  3-­‐(4,5-­‐dimetiltiazol-­‐2-­‐il)-­‐2,5-­‐ difeniltetrazolio  

NLC  

Nanostructured  lipid  carriers  

Transportadores  lipídicos  nanoestructurados  

O/W  

Oil-­‐in-­‐water  simple  emulsion  

Emulsión  simple  orgánico-­‐en-­‐acuoso  

P-­‐gp  

P-­‐glycoprotein  

P-­‐glicoproteína  

PAF  

Platelet  activating  factor  

Factor  activador  de  plaquetas  

PBL  

Peripheral  blood  lymphocytes  

Linfocitos  de  sangre  periférica  

PBS  

Phosphate-­‐buffered  saline  

Tampón  fosfato  salino  

iv  

  Abbreviations  /  Abreviaturas    

PC  

Phosphatidyl  choline  

Fosfatidilcolina  

PCS  

Photon  correlation  spectroscopy  

Espectroscopía  de  correlación  fotónica  

PDI  

Polydispersity  index  

Indice  de  polidispersión  

PEG  

Polyethylene  glycol  

Polietilen  glicol  

PLA  

Polylactic  acid  

Ácido  poliláctico  

PLGA  

Polylactic  co-­‐glycolic  acid  

Ácido  poliláctico  co-­‐glicólico  

PMS  

N-­‐methyl-­‐dibenzopyrazine   methylsulphate  

Metilsulfato  de  N-­‐metil-­‐dibenzopirazina  

PP  

Peyer's  patches  

Placas  de  Peyer  

QC  

Quality  control  

Control  de  calidad  

QD  

Quantum  dot  

Quantum  dot  

RES  

Reticuloendothelial  system  

Sistema  reticuloendotelial  

RSD  

Relative  standard  deviation  

Desviación  estándar  relativa  

S/N  

Signal-­‐to-­‐noise  ratio  

Ratio  señal/ruido  

SA  

Stearyl  amine  

Estearil  amina  

SCID  

Severe  combined  immunodefficiency  

Inmunodeficiencia  severa  combinada  

SIM  

Selected  ionization  monitoring  

Monitorización  de  ionización  seleccionada  

SLN  

Solid  lipid  nanoparticles  

Nanopartículas  lipídicas  sólidas  

t½Ka  

Absorption  half  life  

Semivida  de  absorción  

t½α  

Distribution  half  life  

Semivida  de  distribución  

t½β  

Elimination  half  life  

Semivida  de  eliminación  

TLC  

Thin  layer  chromatography  

Cromatografía  en  capa  fina  

Tmax  

Time  to  peak  concentration  

Tiempo  requerido  para  alcanzar  la   concetración  máxima  

TRAIL  

Tumor  necrosis  factor-­‐related   apoptosis-­‐inducing  ligand  

Ligando  inductor  de  apoptosis  asociado  a   factor  de  necrosis  tumoral  

UHPLC-­‐ MS/MS  

Ultra  high  performance  liquid   cromatography  tandem  mass   spectrometry  

Cromatografía  líquida  ultrarrápida  acoplada  a   espectrometría  de  masas  en  tandem  

VEGF  

Vascular  endothelial  growth  factor  

Factor  de  crecimiento  endotelial  vascular  

Vss  

Volume  of  distribution  

Volumen  de  distribución  

W/O  

Water-­‐in-­‐oil  simple  emulsion  

Emulsión  simple  acuoso-­‐en-­‐orgánico  

W/O/W  

Water-­‐in-­‐oil-­‐in  water  multiple   emulsion  

Emulsión  múltiple  acuoso-­‐en-­‐orgánico-­‐en-­‐ acuoso  

WHO  

World  Health  Organization  

Organización  Mundial  de  la  Salud  

v  

 

 

 

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

   

Introduction    

Lipid  nanomedicines  for  anticancer  drug  therapy   Ander  Estella-­‐Hermoso  de  Mendoza1,  Miguel  A.  Campanero2,  Faustino  Mollinedo3,   María  J.  Blanco-­‐Prieto1     1Departamento  de  Farmacia  y  Tecnología  Farmacéutica,  Facultad  de  Farmacia,  

University  of  Navarra,  E-­‐31008,  Spain   2Servicio  de  Farmacología  Clínica,  Clínica  Universitaria,  E-­‐31080  Pamplona,  Spain   3Centro  de  Investigación  del  Cáncer,  Instituto  de  Biología  Molecular  y  Celular  del  

Cáncer,  CSIC-­‐Universidad  de  Salamanca,  Campus  Miguel  de  Unamuno,  E-­‐37007   Salamanca,  Spain             Journal  of  Biomedical  Nanotechnology,  2009  (5)  p.  323-­‐343  

1  

 

 

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Copyright © 2009 American Scientific Publishers All rights reserved Printed in the United States of America

Journal of Biomedical Nanotechnology Vol. 5, 1–21, 2009

Lipid Nanomedicines for Anticancer Drug Therapy Ander Estella-Hermoso de Mendoza1 , Miguel A. Campanero2 , Faustino Mollinedo3 , and María J. Blanco-Prieto1!∗ 1

Departamento de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, University of Navarra, E-31008, Spain 2 Servicio de Farmacología Clínica, Clínica Universitaria, E-31080 Pamplona, Spain 3 Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain

REVIEW

More than half of all people diagnosed with cancer receive chemotherapy. Unfortunately, most chemotherapy drugs cannot tell the difference between a cancer cell and a healthy cell. In this sense, some other drawbacks often encountered with antineoplastic compounds, such as poor stability and specificity and a high occurrence of drug-resistant tumor cells may be overcome to some degree by incorporating them into drug delivery systems. Solid Lipid Nanoparticles (SLN) have arisen considerable interest in recent years. These are particles of submicron size made from a lipid matrix that is solid at room and body temperature. Moreover, the biodegradable and biocompatible nature of SLN makes them less toxic than other nanoparticulate systems. SLN are capable of encapsulating hydrophobic and hydrophilic drugs, and they also provide protection against chemical, photochemical or oxidative degradation of drugs, as well as the possibility of a sustained release of the incorporated drugs. Along with these last issues, the feasibility of scaling up for large scale production and the low cost of lipids as compared to biodegradable polymers or phospholipids have favoured their use as potential drug delivery systems. This review focuses on the recent advances in SLN as carriers for chemotherapeutic agent delivery

Keywords: Solid Lipid Nanoparticles, SLN, Nanomedicines, Cancer, Antineoplastic.

CONTENTS 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2. Lipid Based Nanosystems . . . . . . . . . . . . . . . . . . . . . . . . 2.1. Solid Lipid Nanoparticles . . . . . . . . . . . . . . . . . . . . . 2.2. Nanostructured Lipid Carriers (NLC) . . . . . . . . . . . . . . 2.3. Lipid Drug Conjugates (LDC) . . . . . . . . . . . . . . . . . . 2.4. Surface Modified Lipid Based Nanosystems: Active Targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . 3. Preparation Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. High Pressure Homogenization (Hot and Cold) . . . . . . . 3.2. Microemulsion Technique . . . . . . . . . . . . . . . . . . . . . 3.3. Emulsion Formation Solvent-Evaporation or -Diffusion Method . . . . . . . . . . . . . . . . . . . . . . . . . . 3.4. Water-in-Oil-in-Water (w/o/w) Double Emulsion Method . . . . . . . . . . . . . . . . . . . . . 3.5. Emulsification Dispersion Followed by Ultrasonication . . 3.6. Hot Homogenization by High Shear Homogenization and/or Ultrasonication . . . . . . . . . . . . . . . . . . . . . . . 3.7. Solvent Injection Method . . . . . . . . . . . . . . . . . . . . . 4. Characterization of SLN . . . . . . . . . . . . . . . . . . . . . . . . . 4.1. Particle Size . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2. Surface Charge—Zeta Potential . . . . . . . . . . . . . . . . . 4.3. Atomic Force Microscopy . . . . . . . . . . . . . . . . . . . . . 4.4. Crystallinity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ∗

Author to whom correspondence should be addressed.

J. Biomed. Nanotechnol. 2009, Vol. 5, No. 4

1 3 3 6 6 6 7 7 7 7 8 8 8 8 8 8 9 9 9

4.5. Stability . . . . . . . . . . . . . . . . . . . . . . . . . 4.6. Lyophilization . . . . . . . . . . . . . . . . . . . . . 5. Efficacy and Toxicity Studies of SLN Loaded with Cytotoxic Drugs . . . . . . . . . . . . . . . . . . . . . . . 6. Pharmacokinetics and Tissue Biodistribution of SLN 6.1. General Aspects . . . . . . . . . . . . . . . . . . . . 6.2. Routes of Administration . . . . . . . . . . . . . . 6.3. Pharmacokinetics . . . . . . . . . . . . . . . . . . . 6.4. Tissue Distribution . . . . . . . . . . . . . . . . . . 7. Concluding Remarks . . . . . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . . . . . Reference and Notes . . . . . . . . . . . . . . . . . . . .

....... .......

9 10

. . . . . . . . .

10 12 12 13 15 16 18 18 18

. . . . . . . . .

. . . . . . . . .

. . . . . . . . .

. . . . . . . . .

. . . . . . . . .

. . . . . . . . .

1. INTRODUCTION Cytotoxic drugs remain the mainstay of cancer chemotherapy.1 The effective use of cancer chemotherapy requires a thorough understanding of the principles of neoplastic cell growth kinetics, basic pharmacologic mechanisms of drug action, pharmacokinetic and pharmacodynamic variability, and mechanisms of drug resistance. Recent scientific advances in the field of molecular oncology have led to the identification of large numbers of potential targets for novel anticancer therapies. Hence, from a pharmacological point of view, these drugs present

1550-7033/2009/5/001/021

doi:10.1166/jbn.2009.1042

1

  3  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy

different mechanisms of action which make them appropriate for one type of cancer or another. Some anticancer agents induce their cytotoxic effects during specific phases of the cell cycle, whereas others show non-specific cytotoxic activity. Antimetabolites, such as fluorouracil (5-FU) and methotrexate, are more active against S-phase cells, whereas the vinca alkaloids and taxanes are relatively more M-phase specific. These differences in

Estella-Hermoso de Mendoza et al.et al. Mendoza

the mechanism of action may have clinically important consequences for cancer chemotherapy. For example, cellcycle–nonspecific agents, such as the alkylating agents and platinum derivatives, generally have linear dose-response curves (e.g., increasing the dose increases cytotoxicity). In contrast, cell-cycle–specific agents will often plateau in their concentration-dependent effects because only a subset of proliferating cells remain fully sensitive to

REVIEW

Ander Estella-Hermoso de Mendoza was got his Bachelor of Science in Pharmacy (2005) and his Master of Science in Pharmaceutical Technology (2007) at the University of Navarra. He is currently a Ph.D. student at the Department of Pharmacy and Pharmaceutical Technology in the same University. His research studies are focused on the development, in vitro and in vivo characterization of polymer and lipid based drug delivery systems containing alkyl lysophospholipid antineoplastic drugs.

Miguel A. Campanero studied Pharmacy at the University of Navarra and received his Ph.D. at the same University in 1998. He is associate professor of Pharmacology at the Faculty of Medicine and Head of the Pharmacokinetics Laboratory from the Clinical Investigation Unit of the University of Navarra. His current research field is preclinical and clinical evaluation of drug delivery systems, covering aspects of bioanalytical evaluation and pharmacokinetic/pharmacodynamic characterization of drug distribution profiles.

Faustino Mollinedo studied Chemistry at the Complutense University of Madrid (Spain), where he obtained his Ph.D. in 1982. After postdocs in Dartmouth Medical School (Hanover, NH, USA), and the New York University Medical Center (NY, USA) (1982–1985), he became a group leader in 1986 in the Center of Biological Research (CIB, CSIC) in Madrid (Spain). In 1994 he moved as group leader and Main Investigator to the Institute of Biology and Molecular Genetics (University of Valladolid-CSIC9 in Valladolid (Spain) (1994–2000), and then to the Center for Cancer Research (University of Salamanca-CSIC) of Salamanca (Spain) (2000-present), becoming Full Professor in 2002. His current work is mainly focused on phospholipid ethers, selective killing of tumor cells, apoptosis and cancer, lipid rafts in apoptosis, and neutrophil differentiation, activation and apoptosis. María J. Blanco-Prieto received her Pharmacy Degree from the University of Santiago de Compostela (Spain) in 1991, followed by a Ph.D. in Pharmaceutical Sciences (1996) from the University of Paris-Sud (France). From 1996 to 1999 she completed a post-doctoral training at the Swiss Federal Institute of Technology (ETH), Zürich, (Switzerland) and then joined the University of Navarra as research scientist and associate professor. Since January 2006, she is the director of the line of research “Nanotechnologies and Drug Delivery Systems” at the Department of Pharmacy and Pharmaceutical technology, University of Navarra. Her research lay in the field of biomaterials and advanced drug carrier systems including the design and the development of polymer and lipid based micro- and nanoscale carriers, their biological evaluation in in vitro cell cultures (toxicity, mechanism of action, intracellular drug release) and also their pharmacokinetic and dynamic impact in vivo (using relevant animal models of the diseases). Research carried out in this field is mainly applied to cancer, infectious, cardiovascular and neurodegenerative diseases. 2

J. Biomed. Nanotechnol. 5, 1–21, 2009

  4  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy

Mendoza et al. de Mendoza et al. Estella-Hermoso

Table I.

Classification of antineoplastic agents vectorized in SLN.

Group Alkaloids Alkylating agents Alkyl Lysophopholipids Anthracyclines Antimetabolites Hormonal agents Podophyllotoxin derivatives Taxanes Others

Examples Mitotic inhibitor: Vinorelbine Topoisomerase I inhibitor: Camptothecin Nitrogen mustard: Chlorambucil Edelfosine Doxorubicin, Idarubicin Folic acid analog: Methotrexate Pyrimidine analog: 5–Fluorouracil Estrogen receptor antagonist: Tamoxifen Etoposide Paclitaxel, Docetaxel All trans retinoic acid Beta–elemene Cholesteryl butirate Cisplatin Topoisomerase II inhibitor: Mitoxantrone

J. Biomed. Nanotechnol. 5, 1–21, 2009

route of administration for all these cytotoxic drugs is the intravenous bolus or infusion, classically in the form of free drug solutions, and even if they present a very long history of use and new drug regimes have been developed in order to improve their clinical accomplishment, it is still frequent to encounter treatment failures.1! 15 This review focuses on the recent advances in SLN as carriers for the delivery of chemotherapeutic agents listed in Table I.

2. LIPID BASED NANOSYSTEMS SLN, NLC and LDC have been designed to overcome the main problems of membrane stability and drug leakage associated with liposomes and conventional emulsions.16 Table II summarizes the antineoplastic drug-loaded lipid formulations developed so far. 2.1. Solid Lipid Nanoparticles SLN are a relatively new class of drug carriers. It was in the mid-1980s when Speiser developed the first micro and nanoparticles (named nanopellets), made of solid lipids for oral delivery.17 Since then, lipids have turned out to be attractive materials for drug delivery system preparation. SLN are characterized as being colloidal particles made of lipids that remain in solid state at room and body temperature, which are generally recognised as safe (GRAS) or have a regulatory accepted status. They present sizes from 50 to 500 nm, depending on the method and materials employed for the manufacturing process. They were introduced by Gasco in the early 1990s, produced by the dilution of a warm microemulsion,18 and by Müller et al.,19 who produced them by a high pressure homogenization method. Advantages that present SLN are the use of biodegradable physiological lipids, the bioavailability improvement of poorly water-soluble molecules, enhanced drug penetration into the skin after topical administration and protection of chemically labile agents from degradation in the gut.20–23 Different stabilizing agents or surfactants confer various interesting properties on the systems. For instance, the use of steric stabilizers such as polysorbates or poloxamers hinders the anchoring of the lipase/colipase complexes in the gastrointestinal tract and consequently, the degradation of the SLN.24 Besides, the use of Tween 80 or sodium dodecylsulfate can make SLN cross the blood brain barrier and deliver drugs into the brain.25–28 Some limitations to these SLN are the likely expulsion of the encapsulated drug during storage and relatively low drug loading. This expulsion phenomenon can occur along the time-dependent restructuring process that happens during storage, which alters the crystallization pattern of the lipid in which the drug is embedded. The step from an imperfect crystalline lattice of the lipid to a more perfect lipid crystalline structure leads to drug expulsion.29–31

REVIEW

drug-induced cytotoxicity. These cell-cycle-specific agents tend to be schedule dependent, because the only way to increase the total cell kill is by extending the duration of exposure, not by increasing the dose.2 In this sense, lipid based particulate systems offer great promise as drug carriers to improve the therapeutic effectiveness and safety profile of these conventional forms of chemotherapeutic agents, by providing a sustained release of the drug over time.3–7 All these DNA-affecting drugs treat cancer principally by being toxic to cells that grow and divide very rapidly. Unfortunately, most of these drugs also affect non cancerous cells, leading to undesirable adverse effects. However, new synthetic ether-linked analogues of phosphatidylcholine and lysophosphatidylcholine, collectively named as antitumour lipids (ATLs), have been developed.8–10 They were initially synthesized in the late 1960s, but have attracted greater interest since the finding that the prototype molecule of the group, the ether lipid 1-O-octadecyl-2-O-methyl-rac-glycero3-phosphocholine (ET-18-OCH3 , edelfosine), selectively accumulates in tumor tissue11 and induces a selective apoptotic response in tumour cells, sparing normal cells. Unlike most chemotherapeutic agents currently used, edelfosine does not interact with DNA, but acts at the cell membrane, and therefore its effects seem to be independent of the proliferative state of target cells.12 Recent progress has unveiled the molecular mechanism underlying the apoptotic action of edelfosine, involving membrane rafts and Fas/CD95 death receptor,13 and has led to the proposal of a two-step model for the selective action of edelfosine on cancer cells, namely: (a) edelfosine uptake into the tumour cell, but not into normal cells; (b) intracellular activation of Fas/CD95 through translocation and capping into membrane rafts. Edelfosine constitutes the first antitumour drug acting through the intracellular activation of the Fas/CD95 death receptor.9 However, drawbacks such as poor oral bioavailability and hemolytic toxicity have been described for this drug.11! 14 The main

3

  5  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy Table II.

Overview of antineoplastic agents vectorized in lipid nanoparticles, composition and preparation methods.

Drug

Lipid matrix

Surfactants/ligands

5-fluorouracil

Triolein, cholesterol

All trans retinoic acid

Tricaprin

Beta-elemene Camptothecin

Monostearin, Precirol Precirol Compritol Precirol, squalene Stearic acid

Hydrogenated soybean phosphatidylcholine, DSPE, Tween 80, ferritin Egg phosphatidyl choline, Tween 80, DSPE-PEG Pluronic F68, Myverol, Pluronic F68 Myverol, Pluronic F68 Myverol, Pluronic F68 Soybean lecithin, Pluronic F68

Cisplatin

Monostearin

Tween 80, Pluronic F68

Chlorambucil

Stearic acid

Cetylpalmitate

Pluronic F68, sodium glycocholate Tyloxapol, sodium glycocholate Pluronic F68, sodium glycocholate, PEG2000 -DSPE Pluronic F68, Tween 60, DDAB Epikuron 200, sodium taurocholate, butanol Tween 80, Span 85, DOTAP

Precirol

Tween 80, DOTAP

Docetaxel

Trimyristin

Doxorubicin

Stearic acid

Egg phosphatidyl choline, galactosylated DOPE Epikuron 200, sodium taurocholate Epikuron 200, sodium taurocholate, PEG2000 Tween 80

Stearic acid, oleic acid

REVIEW

Estella-Hermoso de Mendoza et al.et al. Mendoza

Stearic acid Stearic acid Cholesteryl butirate DNA

Stearic acid Edelfosine

Compritol, stearic acid

Monostearin

Etoposide

Stearic acid

Hydrogenated soybean phosphatidylcholine, sodium tauroglycocholate Hydrogenated soybean phosphatidylcholine, sodium tauroglycocholate Hydrogenated soybean phosphatidylcholine, sodium tauroglycocholate Epikuron 200, sodium taurocholate Soybean lecithin, sodium taurodeoxycholate Soybean lecithin

Monostearin

Soybean lecithin

Tristearin

Soybean lecithin

Compritol

Soybean lecithin

Compritol

Lecithin, polyoxyl-40-stearate

Distearin

Tripalmitin

Idarubicin

Stearic acid

Methotrexate

Stearic acid

Mitoxantrone

Preparation method

% EE

Reference

Solvent injection method

36.8

39

High pressure homogenization Hot homogenization Hot homogenization Hot homogenization Hot homogenization High pressure homogenization Emulsification dispersion– ultrasonication method Hot homogenization

78.7

57

97–99 n.d.

99.6

78 35 35 35 58

82.3

66

>95

67

Hot homogenization

67

Hot homogenization

67

Hot homogenization

67

Microemulsion technique

n.d.

6

High pressure homogenization Emulsion formation solvent evaporation method Hot homogenization

>90

63–65

n.d.

74

>90

68

99

69, 72

Microemulsion technique Microemulsion technique Emulsion formation solvent evaporation method High pressure homogenization

4, 70 85

30

>95

59

High pressure homogenization

59

High pressure homogenization

59, 60

Microemulsion technique Hot homogenization Emulsion formation solvent diffusion method Emulsion formation solvent diffusion method Emulsion formation solvent diffusion method Emulsion formation solvent diffusion method Emulsification dispersion– ultrasonication method

98

71

51.3

16

55–75

75 75 75 75

87.3

5

continued

4

J. Biomed. Nanotechnol. 5, 1–21, 2009

  6  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy

Estella-Hermoso Mendoza et al. de Mendoza et al. Table II.

Continued Lipid matrix

Paclitaxel

Tristearin

Pluronic F68

Stearic acid

Pluronic F68

Compritol

Pluronic F68

Monostearin

Pluronic F68, PEG

Monostearin

Pluronic F68, Folic acid

Trimyristin

Egg phosphatidyl choline, PEG2000 -DSPE Epikuron 200, cholesteryl hemisuccinate, sodium taurocholate, butanol Epikuron 200, sodium taurocholate Lecithin, oleic acid, Pluronic F68

Tripalmitin

Tamoxifen

Palmitic acid

Vinorelbine bitartrate

Monostearin

Surfactants/ligands

Preparation method

% EE

Reference

Emulsion formation solvent diffusion method Emulsion formation solvent diffusion method Emulsion formation solvent diffusion method Emulsion formation solvent diffusion method Emulsion formation solvent diffusion method High pressure homogenizaton Microemulsion technique

36.2

48

59–77

48

Microemulsion technique High pressure homogenization

48 48 48 72–89

61

n.d.

95

28–31

3

80

62

Abbreviations: DOPE—Dioleoylphosphatidyl ethanolamine, DDAB—Dimethyl distearyl ammonium bromide, DSPE—Distearoylphosphatidyl ethanolamine, DOTAP— dioleoyl trimethylammonium propane, PEG2000 —Polyethylene glycol 2000, n.d.—Not determined.

Therefore, the incorporation capability of drugs depends not only on physical-chemical properties of the drug but also on the miscibility, solubility of the drug in the lipid melt, type of matrix material and the degree of crystallinity and polymorphic form.23! 32 Drug molecules that

are not accommodated within the crystalline lattice may adsorb onto nanoparticle surface. There are different models described in the literature to explain the mechanisms of drug incorporation (Fig. 1(A)). The major work has been performed by the research group of Mehnert and

REVIEW

Drug

(A)

Drug-enriched core

Drug-enriched shell

Homogeneous matrix or solid solution

(B)

Fig. 1.

(A) Drug incorporation models for the three types of SLN and (B) basic structure of NLC.

J. Biomed. Nanotechnol. 5, 1–21, 2009

5

  7  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

REVIEW

Lipid Nanomedicines for Anticancer Drug Therapy

co-workers.21! 33 The drug-enriched core is formed when cooling of the hot o/w emulsion leads to precipitation of the drug first. This happens preferentially in lipid solutions with drug dissolved at its saturation solubility in the lipid at production temperature. During cooling, super saturation and subsequent drug precipitation are obtained. The drug-enriched shell model can be explained as a function of the solubility of the drug in the water-surfactant mixture at increased temperature during the production process. The drug partially leaves the lipid particle and dissolves in the aqueous phase during hot homogenization. Cooling of the oil/water nanoemulsion reduces the drug solubility in the aqueous phase, and the drug tries to distribute into the lipid particles leading to enrichment in the particle shell if the particle core has already started to solidify. In the case of a solid solution, the drug is molecularly dispersed uniformly in the particle matrix. Drug release takes place by diffusion from the solid lipid matrix and additionally by further lipid nanoparticle degradation.34 2.2. Nanostructured Lipid Carriers (NLC) The aforementioned problems of drug loading with SLN can be minimized by the recently developed NLC (Fig. 1(B)) by the creation of a lipid particle matrix as imperfect as possible in order to accommodate the active molecules. To achieve this aim, a blend of solid lipid and liquid lipid is used to produce nanoparticles that remain solid at temperatures up to 40 ! C. NLC present considerable crystal disorder, translated into a higher drug loading and less drug expulsion during storage.35 Nevertheless, it has been observed that the controlled release properties of the carrier may be compromised due to the decrease in diffusion length of the lipid matrix.36 However, this drawback can be modulated by altering the proportions of solid and liquid lipids.23 Upon administration of SLN and NLC via the parenteral route of administration, improved biodistribution profile, targeting, and enhanced cytotoxicity against multidrug resistant cancer cells have been observed.35! 37 2.3. Lipid Drug Conjugates (LDC) Due to their lipophilic nature, SLN and NLC present a great advantage for highly efficient incorporation of hydrophobic drugs, but with only a moderate loading capacity for hydrophilic compounds.34 Therefore, in order to solve this problem LDC were developed. In this case, the hydrophilic drug is transformed into a more hydrophobic molecule by conjugation with a lipid compound. Additionally, LDC present advantages for oral and parenteral administration. After oral administration, the bioavailability will be enhanced in comparison to the administration of micronized conjugate powder. After intravenous (i.v.) administration, the LDC nanoparticles offer improved possibilities for drug targeting and distribution through 6

Estella-Hermoso de Mendoza et al.et al. Mendoza

the organism according to the partition coefficient of the drug.38 LDC can be prepared by salt formation or alternatively by covalent linkage. Most of the lipid conjugates melt around 50–100 ! C. These conjugates are melted and dispersed in a hot stabilizing agent solution. Further processing is performed equal to SLN and NLC. The obtained emulsion system is cooled, and the conjugate recrystallises and forms LDC nanoparticles.38 This LDC suspension could to some extent be considered as a nanosuspension of a pro-drug. 2.4. Surface Modified Lipid Based Nanosystems: Active Targeting The in vivo fate of SLN can be customized by a surface modification procedure. This goal can be easily accomplished by the use of ligands that specifically bind to surface receptors on target sites.26! 39 In the case of cancer, there are many receptors reported in the literature that seem to be overexpressed in neoplastic cells. Some examples are folic acid or ferritin receptors (overexpressed in carcinomas),39–42 peptide receptors (growth factors, gastrin related peptides, vasoactive intestinal peptide)43–45 and receptors for LDL (in breast cancer, colon cancer).46! 47 The attachment of suitable ligands on to the surface of lipid nanoparticles results in increased selectivity for tumor tissue.48–52 But the antigen that will allow molecular targeting must be expressed exclusively on cancer cells, be an integral part of an essential cellular function of the cancer cells, and should not easily mutate as the cancer cells proliferate.53 Khalid et al.52 observed the increase in the biological half-life while providing substantial accumulation at the tumour site after the administration of poly(ethylene glycol)-coated SLN to subcutaneously implanted C26 colon adenocarcinoma bearing mice. Stella et al.54 exploited folic acid as a targeting agent to actively target tumor cells, especially in ovarian cancer, while other authors showed that the folic acid-coated lipid nanoparticles containing paclitaxel could enhance the cellular uptake of SLN and the cellular cytotoxicity of the drug by the improved endocytosis mediated by folate receptor in A549 lung cancer cells.48 Stevens et al.51 demonstrated that the treatment of mice bearing lung carcinoma M109 tumours with folate receptor-targeted SLN containing the same drug resulted in significantly greater tumour growth inhibition and animal survival compared to treatment with nontargeted SLN or free paclitaxel formulated in Cremophor EL. Similarly, Jain et al.39 administered 5-fluorouracil (5-FU), SLN and ferritin-coated SLNs to MDA-MB-468 tumour bearing mice. The administration of ferritin-coated SLN formulation also resulted in effective reduction in tumour growth as compared with free 5-FU and plain SLN. Intact antibodies or their fragments have also been used as highly specific targeting agents. Beduneau et al.55 conjugated a whole monoclonal antibody to LNC directed J. Biomed. Nanotechnol. 5, 1–21, 2009

  8  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso Mendoza et al. de Mendoza et al.

against transferrin receptors, enhancing the uptake of these particles by tissues overexpressing such receptor. Recently, delivery of actives to different tissues using nanoparticulate drug carriers in combination with novel targeting principles of “differential protein adsorption (PathFinder Technology)” has been studied in depth.28! 38! 56 This technology identifies the naturally occurring mechanism for localization of material in different parts of the body via adsorbed blood proteins. The production of particles with appropriate surface properties leads in vivo to preferential adsorption of the targeting protein and subsequently to site-specific i.v. delivery.38

3. PREPARATION METHODS

Table III. Main components employed in the formulation of SLN in the field of cancer. Lipid matrix

Caprylic/capryc triglyceride (Mygliol 812) Cetylpalmitate Cholesterol Glyceryl trilaurate (Dynasan 112) Glyceryl trimyristate (Dynasan 114) Glyceryl tripalmitate (Dynasan 116) Glyceryl tristearate (Dynasan 118) Glyceryl monostearate (Imwitor 900) Glyceryl behenate (Compritol 888 ATO) Oleic acid Precirol ATO 5 Solid paraffin Stearic acid

Surfactants

Phosphatidyl choline (Epikuron) Lecithins (egg, soy) Pluronic F68 Polysorbate 80 (Tween 80) Solutol HS15

Co-surfactants

Sodium glycocholate Sodium taurocholate Sodium tauroglycocholate Sodium taurodeoxycholate Cholesteryl hemisuccinate

Cryoprotectants

Trehalose Sucrose Sorbitol Maltose Glucose Mannose

Molecules on surface

Polyethylene glicol Folic acid

J. Biomed. Nanotechnol. 5, 1–21, 2009

3.1. High Pressure Homogenization (Hot and Cold) Patented by Müller and Lucks,19 the high pressure homogenization is a method used to achieve particles with narrow distribution in size. In hot high pressure homogenization, drugs are usually dispersed or dissolved in a molten solid lipid, normally 5–10 ! C above the melting point of the lipid. Next, the drug-lipid melt is dispersed in a surfactant solution kept at the same temperature by high-speed stirring, yielding an aqueous o/w primary emulsion. This pre-emulsion is passed through a high pressure homogenizer, at the same temperature as the previous step. Classic homogenization parameters for this method are from one to three homogenization cycles of 500 bar. The more cycles that are performed, the greater the reduction in size and polidispersity index obtained.22! 34 In the case of the cold homogenization process, the drug is dissolved in the melted lipid followed by a fast cooling by means of dry ice or liquid nitrogen, which leads to the formation of a solid solution. This solution is then micronized with the aid of a mortar or a ball mill, and those solid lipid microparticles are then dispersed and emulsified in a cool aqueous phase containing surfactants. This method overcomes most formulation problems encountered with thermolabile drugs. All-trans retinoic acid,57 camptothecin,58 etoposide,59! 60 paclitaxel,61 vinorelbine bitartrate62 or DNA63–65 are some examples of drugs incorporated into lipid nanosystems by high pressure homogenization.

REVIEW

SLN can be obtained in many ways, from the methods in which organic solvents are employed to more modern methods incorporating the use of high pressure homogenizers. The great variety of materials (solid lipids, liquid lipids, surfactants) and homogenizing techniques can lead us to the achievement of the multiple lipid systems described previously, each one presenting its own physicalchemical profile. Table III summarizes the most widely used excipients in the formulation of antineoplastic drug loaded SLN.

Lipid Nanomedicines for Anticancer Drug Therapy

3.2. Microemulsion Technique The method developed by Gasco et al.18 has been adapted or modified by many authors according to their needs.3! 16! 35! 39! 48! 52! 57! 60! 61! 66–71 Briefly, this method can be explained as the preparation of a warm microemulsion by stirring, containing typically around 10% molten solid lipid, 15% surfactant and up to 10% cosurfactant. This warm microemulsion is then dispersed under stirring in excess cold water (typical ratio 1:50) using a specially developed thermostated syringe. The excess water is removed either by ultra-filtration or by lyophilization in order to increase the particle concentration. Drugs encapsulated by means of this technique are cholesteryl butirate,6 tamoxifen,3 paclitaxel,6 idarubicin71 and doxorubicin.4! 69! 70! 72 3.3. Emulsion Formation Solvent-Evaporation or -Diffusion Method In the emulsion formation solvent-evaporation method,5! 30 the lipid is completely dissolved in a water-immiscible organic solvent (e.g., chloroform) which is then emulsified in an aqueous surfactant solution before evaporation of the solvent under reduced pressure. Once the solvent evaporates, the lipid precipitates forming SLN. An important 7

  9  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

REVIEW

Lipid Nanomedicines for Anticancer Drug Therapy

advantage of this method is the avoidance of heat during the preparation, which makes it suitable for the incorporation of highly thermolabile drugs. Drawbacks might arise due to solvent residues in the final suspension. These suspensions are generally quite dilute, because the lipid presents a limited solubility in the organic material. In the end, lipid concentrations in the final SLN dispersion range around 0.1 g/l, and therefore the particle concentration has to be increased by means of ultra-filtration or evaporation. In the solvent-diffusion technique, partially watermiscible solvents (e.g., benzyl alcohol, ethyl formate) are used.73 Initially, they are both saturated with water to make a certain initial thermodynamic balance of both liquids. Later, the lipid is dissolved in the water-saturated solvent and subsequently emulsified with solvent-saturated aqueous surfactant solution at a raised temperature. After the addition of excess water (typical ratio: 1:5–1:10), the SLN precipitate due to diffusion of the organic solvent from the emulsion droplets to the continuous aqueous phase. Similar to the production of SLN via microemulsions, the particle suspension needs to be concentrated by means of ultra-filtration or lyophilization. Mean particle sizes around 100 nm with very narrow size distributions can be achieved by both solvent evaporation methods. Edelfosine 30 and DNA74 are two examples of drugs that have been encapsulated via solvent evaporation method, and paclitaxel 48 or methotrexate,75 examples of drugs incorporated in SLN via solvent diffusion method. 3.4. Water-in-Oil-in-Water (w/o/w) Double Emulsion Method This method consists of the solubilization of the drug in the internal phase of a double emulsion, along with a surfactant capable of preventing the drug from leaching to the external phase during solvent evaporation. In this case, the lipid melt is emulsified with an aqueous drug and surfactant solution at a high temperature by high shear homogenization (e.g., Ultra Turrax). The obtained warm w/o nanoemulsion is the dispersed in the aqueous external phase of the w/o/w emulsion containing a stabilizer under mechanical stirring at 2–3 ! C. The resultant SLN suspension is then concentrated and purified by diafiltration.30! 76! 77 3.5. Emulsification Dispersion Followed by Ultrasonication This simple method is based on the dissolution of the drug, lipid and emulsifier in a common solvent, which is then evaporated under reduced pressure in order to obtain a solvent free drug dispersed lipid phase. This phase is then homogenized with a hot aqueous surfactant solution using a homogenizer, and then ultrasonicated to obtain the nanoemulsion. The SLN are formed upon cooling 8

Mendoza Estella-Hermoso de Mendoza et al.et al.

at room temperature. Cisplatin66 and mitoxantrone5 have been encapsulated following this method. 3.6. Hot Homogenization by High Shear Homogenization and/or Ultrasonication These techniques do not require the involvement of organic solvents or large amounts of surfactants. Molten lipid is added and dispersed in an aqueous solution with the help of high shear homogenization or ultrasonication. The obtained emulsion is left to cool down at room temperature and may be concentrated by ultrafiltration. Even if these techniques are relatively easy and require tools that are readily available in most laboratories, they present some disadvantages such as low dispersion quality, which is often affected by the presence of a small percentage of microparticles. Metal contamination is another major problem presented by ultrasonication. Many antineoplastic drugs have been successfully incorporated into lipid nanocarriers by this method: beta-elemene,78 camptothecin,35 chlorambucil,67 docetaxel68 and methotrexate.16 3.7. Solvent Injection Method This technique is based on the solvent diffusion method. Here, SLN are prepared by rapidly injecting a solid lipid solution in water miscible solvents into water. SLN precipitate due to diffusion of the organic solvent from the emulsion droplets to the continuous aqueous phase. The most widely used solvents in this method are ethanol, acetone, isopropanol and methanol. An example of drug successfully vectorized by this method is 5-fluorouracil.39

4. CHARACTERIZATION OF SLN The physical-chemical characterization of nanoparticles is essential for the development of any formulation, and so for SLN formulations that will be further tested both in vitro and in vivo. 4.1. Particle Size Particle size plays a critical role not only in their clearance by the reticuloendothelial system (RES), but also in the gastrointestinal uptake and in the prevention of embolisms after intravenous administration. For this reason, the precise determination of the particle size is a must. Sizes below 5 "m are required in order not to cause embolisms due to the blocking of the thin capillaries after parenteral administration of lipid nanoparticles.79 On the other hand, sizes no larger than 300 nm are advisable for the intestinal transport to the thoracic duct.80 The most established method for particle size measurement is photon correlation spectroscopy (PCS).81 Nanoparticles are usually polydisperse in nature and polydispersity index (P. I.) gives a J. Biomed. Nanotechnol. 5, 1–21, 2009

  10  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy

Estella-Hermoso Mendoza et al. de Mendoza et al.

measure of size distribution of the nanoparticle population. Therefore, the lower the P. I. value, the more monodispersed a SLN suspension is. For acceptable polydisperse systems some authors affirm that the P. I. should be less than 0.5,26! 82 however, most researchers accept P. I. values below 0.3 as optimum values.30! 83–86 This technique can sometimes lead to erroneous results when there is presence of different particle sizes which may be accidental microbial contamination, particle agglomerates or dust. To avoid these untrue data it is always wise to corroborate the results with other appropriate methods such as electron microscopy or atomic force microscopy (AFM).

(A)

(C)

4.2. Surface Charge—Zeta Potential

4.3. Atomic Force Microscopy AFM is an important tool for studying either particulate or biological samples due to its ability to image surfaces under liquids.88 Currently, this is the only technique available that directly provides structural, mechanical, functional and topographical information about surfaces with nanometer-to angstrom-scale resolution. Figure 2 shows an example of AFM images taken from edelfosine loaded lipid nanoparticles.30 4.4. Crystallinity Differential scanning calorimetry (DSC) and X-ray diffractometry (XRD) are two of the main tools employed to determine the crystallinity and polymorphic behaviour of the components of the SLN. DSC gives information on the melting and crystallization behaviour of all solid and liquid constituents of the particles, while XRD is used to identify specific crystalline compounds, both mineral and organic, based on their crystal structure. In XRD, a powder made up of small crystals is irradiated with a monochromatic beam of X-rays with the assumption that the orientation of the crystallite is random. The beam is diffracted at angles J. Biomed. Nanotechnol. 5, 1–21, 2009

REVIEW

The zeta potential is the overall charge a particle acquires in a specific medium. The significance of zeta potential is that its value can be related to the stability of colloidal dispersions. The zeta potential indicates the degree of repulsion between close and similarly charged particles in dispersion. For particles that are small enough, a high zeta potential will confer stability, i.e., the solution or dispersion will resist aggregation. When the potential is low, attraction exceeds repulsion and the dispersion will break and flocculate. So, colloids with high zeta potential (negative or positive) are electrically stabilized while colloids with low zeta potentials tend to coagulate or flocculate. In general, zeta potentials superior to −60 mV are essential for excellent electrostatic stabilization. Values of −30 mV were enough for good stabilization, hence, indicating good physical stability.87

(B)

Fig. 2. Atomic force microscopy images of freeze-dried Compritol® lipid nanoparticles: multi-particles (A); zoom-in of the selected area of A (B); 3D morphological image (C).

determined by the spacing of the planes in the crystals and the type and arrangement of the atoms. A scanning detector records the diffracted beams as a pattern. The intensity and position of the diffractions are unique to each type of crystalline material. This pattern can be identified, even in complex mixtures, by comparison to reference spectra using a computerized database. XRD investigations have been most important in the elucidation of the manner of arrangement of lipid molecules, their multiplemelting phenomena, phase behaviour and the characterization and identification of the structure of lipid and drug molecules.29! 30 The determination of the cristallinity of the components of the formulation is very important as the incorporated drug may undergo a polymorphic transition. Lipid matrices can also change their polymorphic behaviour leading to a possible undesirable drug expulsion during storage.31 4.5. Stability Unlike polymeric nanoparticles,89 SLN do not tend to swell in the presence of water.75 However, the type of lipid matrices and surfactants, and its concentration may critically affect the stability of formulations. Chemical instability can be assessed by means of decomposition of lipids. In a study of stability of different lipid matrices, Radomska-Soukharev et al.90 concluded that the decomposition of formulations with mono-, di- and 9

  11  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy

triglycerides was around 10% and that the decomposition increased as the concentration of surfactants increased. The formation of a gel is one of the typical physical instabilities of SLN leading to a change in surface area. This gelation takes place by the SLN building up a network and lipid bridges between them.87 Lipid formulations with Imwitor 900 or Dynasan 118 (tristearin) can be named as examples that form gels at temperatures of 40 ! C.90 More recently, Paliwal et al.75 studied the stability of methotrexate-loaded SLN. These authors concluded that 60 days after they were prepared SLN maintained their size at 4 ! C, while this size increased when these particles were kept at 25 ! C.

REVIEW

4.6. Lyophilization Obtaining a dry product in order to ensure stability of SLN is very important. Lyophilization is one of the most used techniques to produce dry powders from nanoparticulate suspensions.91–95 However, parameters employed in this procedure depend on the lipid used for the SLN formulation. Zimmerman et al. optimized the temperature and time parameters for the lipid matrix Imwitor 900 (glyceryl monostearate).79 Firstly, the aqueous SLN-dispersion was diluted with a solution of a cryoprotectant (trehalose or fructose) and frozen at −70 ! C right after production in a deep-freeze. After thermal treatment of 2 hours at −22 ! C and a posterior cooling down to −40 ! C for another 2 hours, formulations underwent a primary drying process at 1.03 mbar (−30 ! C 7 h, −10 ! C 2 h, 20 ! C 12 h). Finally, a secondary drying process for 3 hours at 30 ! C and 0.001 mbar was performed to obtain the lyophilized product. Consequently, the lyophilization process subjects the lipid nanosystems to freezing and drying, two important stresses that can affect their physical stability if suitable stabilizers or cryoprotectants, namely sugars, are not employed. In fact, when SLN are lyophilized without cryoprotectants, the final product commonly results in the aggregation of particles, which acquire a rubbery aspect. The most generally employed cryoprotectants in the formulation of SLN are trehalose, sucrose, sorbitol, maltose, glucose and mannose, among others. In a study of freeze-drying of SLN, Schwarz and Mehnert96 concluded that trehalose was the most effective cryoprotectant in preventing particle growth. Following this line, Del Pozo-Rodríguez et al.74 studied the short- and longterm stability of lyophilized DNA-containing PrecirolSLN cryoprotected with glucose and trehalose. Samples were frozen at −80 ! C. After 24 hours, frozen samples were lyophilized at −55 ! C and 0.2 mbar for 48 hours. 5% glucose-cryprotected SLN rendered a powdered product that became rubber after 48 h, while the use of other glucose concentrations led to rubbery samples from the beginning. In the case of trehalose, a concentration of 10% 10

Estella-Hermoso de Mendoza et al.et al. Mendoza

manifested the best results, yielding a powdered product that remained over the time. This same study showed that the lyophilization process of the SLN meant an increase in the particle size of about 100 nm, whatever the trehalose concentration used. However, this size hardly varied along the first 9 months at room temperature.

5. EFFICACY AND TOXICITY STUDIES OF SLN LOADED WITH CYTOTOXIC DRUGS The local or systemic chemotherapeutic response of a drug may be improved by incorporation of anticancer agents into a delivery system that can facilitate sustained drug release for a sufficient period of time. Therefore, efficacy and therapeutic value of antitumour drug could be substantially diminished by severe systemic toxicity. This is consistent with an in vivo study with Ehrlich ascite carcinoma (EAC) bearing mice performed by Ruckmany et al.16 They administered methotrexateloaded SLN made of stearic acid intraperitoneally for 9 days. After day 9, mean survival time for the mice treated with SLN was nearly 30% higher than that of the mice treated with methotrexate solution. Reddy et al.59 also increased by 5–8 days the mean survival time of mice bearing Dalton’s lymphoma after etoposide-loaded SLN were administered intraperitoneally. SLN were made of glycerol monostearate, glycerol distearate and tripalmitin. Among the three nanoparticle formulations, tripalmitin lipid nanoparticles showed significantly higher apoptosis and increase in survival time of tumour bearing mice. Xu et al.68 observed that docetaxel-loaded galactosylated SLN showed the potential of increasing the efficacy in the treatment of hepatocellular carcinomas expressing the asialoglycoprotein receptor. Furthermore, a SLN-treated group of hepatoma bearing mice demonstrated the most dramatic efficacy of all the tested treatments, and all animals treated showed complete tumour regression with no significant toxicity after a multiple dose administration of docetaxelloaded SLN. In another study, mitoxantrone-loaded SLN also showed efficacy against lymph node metastases. The treatment with SLN gave a mean size of lymph node three times smaller than that of the mitoxantrone solution.5 Table IV shows the most representative in vitro studies performed with antineoplastic agents loaded in SLN. To date, there is no SLN product for cancer therapy on the market. In order to develop a SLN formulation for its administration in clinical practice it is crucial to establish the biocompatibility of the components with blood components and other tissues. For this reason, various in vitro and in vivo studies have been performed to study the toxicity and biocompatibility of lipid nanoparticles. Experiments with the interaction of SLN with human granulocytes revealed that SLN had much lower uptake by the RES, resulting in extended circulation time in blood. The study also showed the nearly 10-fold J. Biomed. Nanotechnol. 5, 1–21, 2009

  12  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy

Estella-Hermoso Mendoza et al. de Mendoza et al. Table IV.

In vitro studies performed with different tumoural cell lines and corresponding IC50 (half maximal inhibitory concentration).

Drug

Lipid matrix

Molecule on surface

Cell line

5-fluorouracil Cholesteryl butirate Docetaxel Doxorubicin

HSPC Epikuron 200 Trimyristin Stearic acid

Ferritin — — —

Paclitaxel

Tristearin Compritol Monostearin Monostearin Trimyristin

— — PEG Folic acid PEG2000 -DSPE

Tamoxifen

Palmitic acid



MDA-MB-648 HT-29 BEL7402 HT-29 U-373 Y-79 HL-60 MCF-7 A549 A549 A549 A549 OVCAR-3 MCF-7 MCF-7

IC50 (!M) free solution

IC50 (!M) SLN

IC50 (!M) targeted SLN

5.02 ≥ 600 — 0.13 2.15 >0.51 0.07 0.02 2.13 2.13 2.13 2.13 ≈ 20 ≈ 0"5 ≈ 100

3.56 300 — 0.08 0.99 0.16 0.001 0.001 0.84 1.33 0.63 0.63 — — ≈ 100

1.28 — — — — — — — — — 1.74 0.19 ≈ 20 0.5—1 —

Reference 39 6 68 69, 72

48 48 48 48 61 3

Abbreviations: DSPE—Distearoylphosphatidyl ethanolamine, HSPC—Hydrogenated soya phosphatidylcholine, PEG2000—Polyethylene glycol 2000, DSPE— Distearoylphosphatidyl ethanolamine, HSPC—Hydrogenated soya phosphatidylcholine, PEG2000—Polyethylene glycol 2000.

J. Biomed. Nanotechnol. 5, 1–21, 2009

an in vitro degradation study.99 In contrast, a lower dose of Compritol SLN (2.5%, w/w) was well tolerated. The binding of SLN formulations to different cells, including red blod cells, is important because it may affect not only blood clotting or embolic effects but also may produce a change in the pharmacokinetic behaviour of the drug. SLN have diverse affinities to erythrocytes depending on the surfactant used. Olbrich et al.24 showed by flow cytometry that SLN formulations (made of Compritol as matrix material and Tween 80 and Poloxamer 188 as surfactants) exhibit a binding to erythrocytes below 10.0%. In contrast, when Span 85 was employed, blood cell affinity of SLN was increased, which translated into a significant aggregation of red blood cells (75.3%). Surfactants are also extremely important components of SLN and their biocompatibility and cytotoxicity must be investigated. Good tolerability depends in the first line on the surfactant used and secondarily on the lipid. To formulate parental SLN, surfactant of GRAS status must be employed, e.g., lecithin, Tween 80, Poloxamer 188, Span 85 or sodium glycocholate. Many authors have studied the toxicity of different stabilizing agents on cells and their capability to reverse multidrug resistance (MDR), suggesting that MDR reversal by polyoxyethylene surfactants involves alterations of the lipids physical state of the plasma membrane in resistant cells as one of the possible mechanism(s).101# 102 Concerning Solutol® HS15, Woodcock et al.102 concluded that 66% of the R100 cells (MDR-derivative of human leukaemia cell line) were lysed when incubated for only 1 h with this polyethoxylated solubilising agent at concentrations equivalent to 1:100 (w/v) and 100% were lysed by concentrations of 1:10 (w/v). This surfactant is greatly employed in the formulation of LNC. Hence, Allard et al.103 investigated the cytotoxicity of a tamoxifen derivative loaded in LNC prepared with

REVIEW

lower cytotoxicity of glyceride SLN compared to that of PLGA nanoparticles.97 As a consequence, the authors concluded that SLN can be used as drug carriers because of their prolonged circulation time and high toxicological acceptance. In an attempt to study the toxicity of lipids employed, SLN made from lipids consisting of stearic acid or dimethyl-dioctadecylammonium bromide were assayed in vitro using murine macrophages in the presence of different concentration and different size of nanoparticles.98 It was concluded that the nanoparticles were cytotoxic at the concentration of 0.01%, independently of the size of the SLN. However, SLN made from lipids consisting of triglycerides, paraffin or cetylpalmitate were found to be harmless at the same concentration. These data agree with the results obtained by Weyhers et al.,99 who in an in vivo study provided a better understanding of the toxicity created by SLN in vivo when two types of SLN, one made from Compritol (GRAS) and the other made from cetyl palmitate (which is less physiologic), were i.v. administered six times within a period of 20 days. The doses that were administered in this study were extremely high, up to 1 g of solid lipid per kg of body weight (10% w/w). Translated into human administration, this could be compared to bolus injection of 75 g of solid particles intravenously.100 The hepatic and splenic tissues were analysed histopathologically. The results showed that the toxicity was dependent not only on the lipid matrix but also on the dose administered. Thus, for the less physiological lipid cetyl palmitate containing SLN no pathological results were obtained, whereas Compritol containing formulations led to a partially reversible accumulation of the lipid in liver and spleen and subsequently to pathological alterations. This accumulation was attributed to the slow degradation of the Compritol matrix compared to the faster degradation of cetyl palmitate that could be observed in

11

  13  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso de Mendoza et al.et al. Mendoza

REVIEW

Lipid Nanomedicines for Anticancer Drug Therapy

Solutol® HS15 using rat 9 L gliosarcoma cells. Tamoxifen derivative-loaded LNC demonstrated cytotoxic activity on 9 L cells 150-fold higher than blank LNC, while cell survival curves were not significantly different between the derivative in solution and loaded in LNC, indicating that the activity of the drug was totally recovered in vitro after encapsulation. The same authors performed an in vivo study using the same tumour cell line-bearing rats, observing that animals treated with a single administration of drug-loaded LNC at an equivalent dose of 2.5 mg/kg presented tumours three times smaller than those treated by blank LNC, indicating that the cytostatic activity of the tamoxifen derivative was preserved in vivo. An in vitro antiproliferative activity of tamoxifen-loaded SLN was also assayed on MCF-7 cell line (human breast cancer cells),3 showing that tamoxifen incorporated in SLN maintained the same antitumour activity as when administered as a free drug. Many in vitro studies performed with doxorubicin loaded SLN have clearly shown the efficacy of the systems.69! 72 In all tested tumour cell lines, doxorubicinloaded SLN inhibited cell growth more strongly than the free solution. Authors could associate the enhanced cytotoxicity with an increased drug accumulation in tumour cells.72 Cytotoxicity of paclitaxel-loaded SLN has also been widely assayed.6! 48! 51! 61! 69! 104 Different authors have discovered that the cellular uptakes of different types of SLN are time-dependent, concentration-dependent and that the intracellular accumulation of drugs may be further optimized through receptor-mediated endocytosis of the drug-loaded SLN, as previously mentioned in this review. These authors determined that the order of cellular uptake ability for the tested SLN was tristearin SLN > monostearin SLN > stearic acid SLN > Compritol ATO888 SLN. Concerning cytotoxicity, IC50 values for paclitaxel demonstrated that monostearin SLN were the less cytotoxic.48 In a another study, Lee et al.61 found no significant differences in cytotoxicity between paclitaxelloaded SLN and the paclitaxel containing Cremophor EL-based formulation, presenting an IC50 value of approximately 20 "M against OVCAR-3 cell line. Compared with MCF-7 cells, the paclitaxel containing Cremophor EL-based formulation had a slightly lower IC50 value than the paclitaxel-loaded SLN, even if this difference was not statistically significant. You et al.62 incorporated vinorelbine bitartrate into SLN and tested the cytotoxicity of the system in a MCF-7 breast cancer cell line. Compared to the free vinorelbine bitartrate, the cellular cytotoxicity of the drug loaded in SLN increased approximately 20% after 24 h in the same incubation conditions, while blank SLN presented a cellular viability of above 80%. 12

6. PHARMACOKINETICS AND TISSUE BIODISTRIBUTION OF SLN 6.1. General Aspects Anticancer drugs are a diverse class of compounds that present specific problems when used in cancer treatment. Poor specificity, high toxicity and susceptibility to induce drug resistance are the main drawbacks that have been observed after their use in humans. Conventionally administered cytotoxic agents extensively and indiscriminately bind to body tissues in a highly unpredictable manner. Only a small fraction of the drugs reaches the tumour site. This may both reduce the therapeutic efficacy and increase systemic drug toxicity. Moreover, even though cytotoxic drugs ideally should only kill cancer cells, in reality they do not differenciate between healthy and cancerous cells. Tumour response is determined by the in vivo drug half-life, the cellular residence time of the drug and the nature of observed cellular responses. Generally, tumours are associated with a defective, leaky vascular architecture as a result of the poorly regulated nature of tumour angiogenesis. Moreover, the interstitial fluid within a tumour is usually inadequately drained by a poorly formed lymphatic system. As a result, submicron-sized particulate matter such as SLN can preferentially extravasate into the tumour and be retained there, maximizing the amount of drug that can reach the targeted tumour sites.7 This phenomenon is known as the enhanced permeability and retention (EPR) effect and is widely exploited in the drug delivery system community. Chemical and physical properties of the nanoparticles, including size, composition, and surface, are also important factors that determine their pharmacokinetics and biodistribution. In general particles, as with all intravenously injected and colloidal particulates, are opsonized and cleared from the circulation by the reticuloendothelial system or mononuclear phagocyte system. To facilitate drug targeting, in tumour tissue for example, a reticuloendothelial system avoidance (stealth) facility may be incorporated as described well for classical polymeric nanoparticels in the past. SLN may be used to target drugs at particular organs without surface modification because of their small size (50–200 nm). Moreover, solid lipid nanoparticles have drawn increasing interest from every branch of medicine owing to their ability to overcome certain biological barriers, resulting in increased therapeutic efficacy of the encapsulated drug. SLN increase the tumour accumulation104 and allow brain delivery of anticancer drugs not capable of crossing the blood brain barrier (BBB).4 Different sub-classes of cytotoxic agents and their derivatives, including anthracyclines (e.g., doxorubicin, idarubicin), taxanes (e.g., paclitaxel), camptothecins (camptothecan, 7-ethyl-10-hydroxy-20(S)-camptothecin (SN38)), etoposide, flurodooxyuridine (FudR) and retinoic acid and other J. Biomed. Nanotechnol. 5, 1–21, 2009

  14  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso Mendoza et al. de Mendoza et al.

investigational compounds (e.g., cholesteryl butyrate) have been encapsulated, and their disposition evaluated in preclinical studies on different species. 6.2. Routes of Administration

6.2.1. Oral Administration of SLN Oral delivery is the easiest and the most convenient route for drug administration, but many drugs are not administered orally because they are not absorbed or are degraded in the gastrointestinal tract. SLN have been proposed to overcome these problems. Absorption of SLN by the gastrointestinal tract has been studied extensively for the last decade. The factors controlling intestinal absorption of particulate systems are now known. Size, nature of the lipid material, inner structure of the vehicle and the employed surfactants have been determined as critical factors influencing particle uptake.98! 106 Reduction in the particle size is a key factor for improving the peroral performance of SLN of poorly soluble drugs. In SLN formulations, the particle size range has been reduced to less than 400 nm, resulting in an increase in surface area and saturation solubility, and making it possible to reach high concentrations of SLN in the gastrointestinal tract.106 Due to their small particle size, SLNs may exhibit bioadhesion properties to the gastrointestinal tract wall or enter the intervillar spaces thus increasing their residence time in the gastrointestinal tract. This increase in adhesion results in enhanced bioavailability. J. Biomed. Nanotechnol. 5, 1–21, 2009

The mechanism of oral absorption for SLN still needs to be revealed. Different mechanisms for the particle absorption from the gastrointestinal tract have been described (Fig. 3). Transport through endothelial tight junctions, endocytosis by the epithelial membranes and M-cell mediated transport have been proposed as possible mechanisms. The main SLN uptake pathway in rats is the M-cells transport overlaying the Peyer’s patches.107 The particle size is a critical determinant of the absorption of nanoparticles administered orally. Larger particles (>300 nm) may be retained for longer periods in Peyer’s patches, while smaller particles are transported to the thoracic duct. Moreover, the composition of SLN can also play a role in relation to the uptake pathway. A wide variety of solid lipids, such as highly purified triglycerides, complex glyceride mixtures, phospholipids or even waxes are employed to prepare SLN. Surfactants are used to stabilize the obtained drug delivery formulation. It is known that liquid lipid vehicles constituted of some unsaturated fatty acids and triglycerides can favour intestinal lymphatic absorption of orally administered drugs. Briefly, fatty acids with chain lengths of 14 or greater are more highly lymphatically transported, whereas shorter chain fatty acids are primarily absorbed via the portal blood. As such, triglycerides composed of long chain fatty acids more effectively support lymphatic drug transport than their medium and short chain counterparts.108 Transport of such drugs or carriers through the intestinal lymphatics, via the thoracic lymph duct to the systemic circulation joining at the junction of the jugular and left subclavian vein, avoids presystemic hepatic metabolism, and as a result, enhances the amount of orally administered drugs reaching into systemic circulation. Several experiments have been performed to ensure the SLN lymphatic uptake after duodenal administration to rats. Transmission electron microscopy has been used to observe the presence of SLN in lymph and blood.107! 109 In some recent research, SLN suspensions were administered by oral gavages to rats with and without thoracic lymph duct cannulation. Approximately 77.9% of absorbed SLN was transported into systematic circulation via lymph, whereas the remaining 22.1% of SLN was absorbed by non-lymphoid pathway.110 The intestinal lymphatic transport of SLN offers some potential advantages. Firstly, direct lymphatic transport bypasses the first pass metabolism in the enterocyte and potentially reduces the access to efflux proteins such as p-glycoprotein, enabling the oral route to be used for the administration of some drugs limited to parenteral route administration. Besides, SLN drug formulations afford the possibility of targeting drugs to the lymph for the treatment of lymphatic cancers. Finally, the lymphatic system acts as a prolonged-release system, affording a sufficiently high level of administered drug even after 24 h.75! 108 In previous research, idarubicin-SLN administered by the duodenal route gave

REVIEW

SLN-anticancer systems are generally injected either intravenously, intraperitoneally or administered orally. The route of administration is an important parameter in the delivery of SLN-encapsulated anticancer agents to tumours. Some hypotheses highlight the ability of SLN to enhance the oral bioavailability of actives by either physical-chemical or biochemical mechanisms. Physical-chemical mechanisms are based on enhancing the solubility of the drug. On the other hand, the biochemical mechanisms base their success in the inhibition of the efflux transporters. Different routes of administration may result in varying effects on the SLN biodistribution pattern. Reddy et al.105 studied the biodistribution and tumour uptake of etoposide-SLN after s.c., i.v. or intraperitoneal administration to tumour bearing BALB/c mice. The obtained results showed that s.c. injection reduced the distribution of etoposide-SLN to all the tissues studied, whereas after intraperitoneal injection, the distribution of etoposide-SLN to tissues was higher. The intravenous injection also resulted in lower concentrations of etoposide nanoparticles in the organs of RES. The influence of the administration route on the bioavailability of SLN must be explained before studying the SLN biodistribution profile and tumour uptake.

Lipid Nanomedicines for Anticancer Drug Therapy

13

  15  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Lipid Nanomedicines for Anticancer Drug Therapy

Estella-Hermoso deMendoza Mendozaetetal. al.

SLN

Adhesion

REVIEW

Anchoring of lipases

Drug release + Lipid degradation products

Degradation drug release

ABSORPTION OF DRUG AND LIPIDS

Micelle, drug solubilized + Bile salts

Mixed micelle

Gut wall Blood

Gut Fig. 3.

Mechanisms of absorption promoting effect of lipids formulated as a lipid nanoparticle.

higher idarubicin levels (around 40%) than those after i.v. administration of the same formulation at 24 h after administration.71 The surfactants contribute to an increase in the permeability of the intestinal membrane or improved the affinity between lipid particles and the intestinal membrane. In previous research,111 the absorption was greatly improved by increasing the amount of Pluronic F68 and Tween 80 on SLN nanoparticles. SLN show a certain physical stability in the gut lumen and lymph ensures a partial protection of the incorporated 14

Mucus

drugs.107 The physical stability of SLN reduces the local adverse effect of some drugs on the gastrointestinal tract, and protects biodegradable molecules, such as peptides and proteins, when orally administered. The encapsulation of anticancer drugs in lipid nanocapsules has also led to an improvement of oral exposure compared to the free drug. The inhibition of the P-glycoprotein by the surfactants contained in the lipid nanocapsules could explain the improvement of oral paclitaxel exposure when it was administered in lipid nanocapsules to male Sprague rats.112 J. Biomed. Nanotechnol. 5, 1–21, 2009

  16  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso Mendoza et al. de Mendoza et al.

6.2.2. Parenteral Administration of SLN

6.2.3. Topical Administration of SLN SLN are considered to be the latest generation of colloidal drug carriers for topical use. Compared with other vehicles, such as creams, tinctures and emulsions, and other delivery systems like liposomes and microparticles, they combine such advantages as local tolerance, good adhesive properties, the protection of the encapsulated drugs against chemical decomposition, and the possibility of modulating the drug release.20 The bioavailability of drugs in nanoparticulate carriers may be increased by the close contact between the small particle size of nanoparticles and stratum corneum. However, the limited voids in the lipid bilayers of stratum corneum make the diffusion of intact particles into the skin impossible. SLN have a more favourable occlusive effect than other pharmaceutical preparations and hence the particle permeation through the skin is increased. SLN form adhesive films of densely packed spheres on the surface of the skin, increasing skin hydration.113 J. Biomed. Nanotechnol. 5, 1–21, 2009

The enhanced skin permeation of the SLN is probably due to the additives included in the formulation. Surfactants, which can loosen or fluidize the lipid bilayers of the stratum corneum, may act as permeation enhancers.20 The biodistribution data after subcutaneous injection (s.c.) injection of etoposide-SLN to tumour bearing BALB/c mice showed that the distribution of encapsulated drug was slow from the injection site. The increase in the concentration of etoposide in tissues at 6 h and 24 h postinjection indicates the etoposide-SLN residence at the injection site for a longer time.105 However, in most studies the evaluation of the observed pharmacological effect is the only procedure to assess the ability of a delivery system in transdermal applications. In vitro measurements can be useful to assert SLN occlusive effects.22 6.2.4. Intratumour Administration of SLN Few studies have been performed to study the effectiveness of SLN after direct intratumour administration. Mitoxantrone is example of drug that after s.c. injection induces more serious local toxicity than intravenous injection or intraperitoneal injection (i.p.). After multiple s.c administration of mitoxantrone loaded SLN every four days, for 28 days, it could be concluded that mitoxantrone-SLN had a strong affinity to the tumour tissue, contributing to the therapeutic effect of the antitumour agent. After mitoxantrone solution s.c. administration, no such affinity to the tumour surface was observed.5

REVIEW

SLN can be used for all parenteral applications suitable for polymeric nanoparticles.22 As mentioned before, the particle size of intravenously administered drug must be below 5 !m to avoid blocking of fine capillaries leading to embolism. Because of their minimum size below 400 nm, SLN formulations can be used for systemic body distribution with a minimized risk of blood clotting and aggregation. Using SLN as carriers, a three to five-fold enhancement of i.v. plasma peaks of the encapsulated drug as well as a prolonged residence time are generally observed after SLN administration. Uptake of the nanoparticles in cells of the RES or mononuclear phagocyte system differed according to the size and composition of the particles. Uptake of SLN can be avoided by PEGylation leading to long circulating particles, so-called stealth SLN. Surprisingly, stealth SLN showed similar circulating time and pharmacokinetic behaviour in comparison to unmodified nanoparticles.4 The similar low surface hydrophobicity of both types of particles could explain the results obtained when stealth and non-stealth doxorubicin SLN were evaluated in vivo. Minimizing opsonin protein binding is the key point for developing a long circulation nanoparticle formulation. The nature of the lipid material and the surfactant employed in the SLN preparation might play an important role to avoid adsorption of blood proteins that mediates liver uptake.23 The incorporated drug is gradually released on erosion (e.g., degradation by enzymes) or by diffusion from the particles. The rate of release may be controlled by the nature of the lipid material, particle size, and choice of surfactant and also by inner structure of SLN34 as discussed above.

Lipid Nanomedicines for Anticancer Drug Therapy

6.3. Pharmacokinetics Administration of anticancer drugs incorporated in SLN leads to different pharmacokinetic profiles to those obtained after free drug administration. When the drugs are encapsulated in SLN, the drugs are protected from chemical degradation in gastrointestinal tract, bypass the effect of extrusion membrane proteins, reduce the drug exposure to hepatic metabolizing enzyme activity and avoid the renal clearance due to increased size. Reduced liver metabolism and renal clearance of drugs encapsulated in SLN often result in prolonged blood circulation, with an increased chance of accumulation in the target tissue. Table V summarizes the key pharmacokinetic parameters in some studies of anticancer agents delivered by SLN. All non-stealth SLN formulations led to significantly higher levels of drug remaining in the systemic circulation for longer periods of time. Using SLN as carriers, mostly a three to twenty-fold enhancement of drug plasma peaks is observed when SLN are administered by i.v. route. The terminal half-lives of the encapsulated drugs are also significantly increased when administered via SLN. Plasma concentration-time profile for SLN shows a bi-exponential curve with high AUC, a lower rate of clearance, and a smaller volume of distribution in comparison to the free drug.107" 109" 110" 114 The biphasic behaviour can probably 15

  17  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso de Mendoza et al.et al. Mendoza

Lipid Nanomedicines for Anticancer Drug Therapy Table V.

Pharmacokinetic parameters of anticancer drugs delivered by lipid nanoformulations. AUC (!g l−1 h)

Animal

Drug

Solution

Non-stealth

Stealth

Solution

Non-stealth

Stealth

References

Mice Rat Rabbit Rat Mice Rat Rat Mice Mice Rat Mice Mice

Camptothecin Doxorubicin Doxorubicin Doxorubicin FuDR Idarubicin (iv) Idarubicin (or) Paclitaxel (or) Paclitaxel (iv) Tamoxifen (iv) Metothrexate (iv) Docetaxel (iv)

66a 5"88 2"86 5"02 0"04 6"51 1"88 0"382 0"624 0"004 1"185 7200

324a 102"8 7"39 48"87 0"138 27"53 40"45 1"05 1"15 0"021 1"987 N.A.

N.A. N.A. 15.6–25.99b 67.27 N.A. N.A. N.A. N.A. N.A. N.A. N.A. 29100

N.A. 2"54 0"80 3"72 2"10 0"79 1"16 N.A. 1"36 2"68 8"2 0"3

0"36 3"36 1"19 4"11 19"0 23"51 7"22 4"5 10"06 9"58 14"46 N.A.

N.A. N.A. 1.20–1.83b 3.52 N.A. N.A. N.A. N.A. N.A. N.A. N.A. 1.4

58 109 4 4 119 71 71 112 104 120 16 52

a

REVIEW

Terminal half-life (h)

Unit of AUC is h ng g−1 . b Stealth SLN with three different levels of stealth agent (PEG2000-stearic acid). N.A. — Not applicable.

be explained by the slow distribution of SLN to organs and tissues. It is interesting to note that sometimes two peaks were observed in the concentration-time profile of encapsulated drug after SLN administration, whichever the administration route was. The first peak of encapsulated drug in blood took place immediately after SLN intravenous administration and for 1–2 h after oral SLN administration which indicated that SLN were absorbed quickly from the gastrointestinal tract into systemic circulation. SLN concentrations then began to decrease, which may be related to the uptake of SLN by the macrophages,115# 116 the distribution of SLN among particular organs and the release of encapsulated drug. The second peak occurred at about 6–8 h, and the maximum concentrations were lower than that of the first drug peak. The second peak of the encapsulated drug may be caused by the re-distribution of SLN released from those particular organs into systemic circulation.110 The interaction of SLN with the major circulatory protein, serum albumin, has been investigated in the present decade. Albumin adsorption on the particle surface formed a capping layer of 17 nm. The SLNs are protected by this layer against flattening on surfaces. At physiological albumin concentrations (35–50 g/l), the increased size was not important enough to explain blood cell aggregation.117 After oral administration of SLN to rats, the bioavailability of the encapsulated drug increased considerably. The AUC values obtained after administration of idarubicin-SLN was higher (around 21-fold) than that of idarubicin free solution.71 The administration route affects the bioavailability of SLN encapsulated drugs strongly. The AUC of SLN of idarubicin after administration into the duodenum was higher than that obtained after SLN i.v. administration.71 The difference between the two administration routes for the most part lies in the predominant transmucosal transport of SLN to the lymph; lymphatic transport predominantly contributes to SLN absorption, which could affect the drug bioavailability. 16

Coating SLN with PEG2000 further improved their abilities to evade the RES clearance.58# 71# 109 The AUC values significantly increased after SLN were coated with the stealth agents (Table V). However, the increase in circulation time was generally not as substantial. The opposite effect was observed when anticancer agents were formulated in lipid nanocapsules. Non-PEGylated docetaxel lipid nanocapsules were rapidly cleared from the systemic circulation, whereas the increase of PEG content from 6 to 10% gave a three-fold elevation of docetaxel blood levels at the same time after intravenous administration on female BALB/c mice inoculated with C26 colon adenocarcinoma cells.52 6.4. Tissue Distribution Like most drugs, anticancer drugs only exercise their therapeutic effect when they diffusses into the tumour. Therefore it is very important to know where exactly an anticancer drug has been distributed. Unfortunately, in vivo efficacy studies of SLN loaded anticancer drugs have so far been rare. Up until now, the study of the biodistribution profile of anticancer drug-loaded SLN in tumour-bearing animals has been considered an adequate tool to predict the therapeutic effect of the loaded drug. The tissue distribution profiles of SLN encapsulated drug differ significantly from the observed profile after administration of the free anticancer drug. With the exception of the brain, concentrations of drugs encapsulated are lower after SLN administration than with the obtained after drug free administration, especially for the liver, spleen, and kidneys, heart and lungs. SLN are widely distributed in muscles, bone, and fatty tissues.58# 108# 114 The reason for this different pattern of body distribution is not so far completely understood. The slower diffusion of SLN through biological barriers and the slower drug release from the SLN are the main reasons that could explain this. J. Biomed. Nanotechnol. 5, 1–21, 2009

  18  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso Mendoza et al. de Mendoza et al.

J. Biomed. Nanotechnol. 5, 1–21, 2009

Stealth SLN increases the brain concentration of the encapsulated drug. Zara et al.71 evaluated the effect of the stealth agent concentration in the brain uptake of doxorubicin SLN in rabbits. They observed an increase in the brain concentration of doxorubicin on increasing the stealth agent. The amount of drug present in the rabbit brain ranged from 27.5 ng/g for nonstealth SLN to 242.0 ng/g for stealth SLN preparation loaded with 0.45% PEG after 30 min. The presence of surfactant agent in the SLN improves its brain uptake. The brain uptake of Poloxamer 188 stabilized stearic acid camptothecin-loaded SLN was studied after both oral and i.v. administration in mice. The aforementioned particles showed a significant brain uptake, and the maximum concentration (Cmax ) increased by 180% when compared to the Cmax of the drug solution.114 Surface charged SLN were also proposed in order to achieve brain targeting. Positively (surface charge: +5 mV) and negatively (surface charge: −47 mV) charged tripalmitin SLN were loaded with labelled etoposide and their brain uptake was compared to that of free labeled drug in mice after i.v. administration. Positively charged SLN showed a higher brain accumulation compared with both negatively charged SLN and free labeled drug. The etoposide concentration, achieved using positively charged SLN, was 10- and 14-fold higher with respect to the negatively charged SLN at 1 and 4 hours after administration, respectively.60 Unfortunately, the strategy of using surface charged SLN to cross the brain blood barrier has shown some important drawbacks such as the toxic effects at brain microvasculature endothelium of some cationic nanoparticles, which limit its usefulness.27

REVIEW

In general, the major limiting factor for the systemic use of nanoparticles is their rapid clearance from the blood circulation by the RES. The physicochemical properties of SLN are responsible for its wide distribution profile. SLN, particularly those in the range of 120–200 nm, are not taken up readily by the cells of the RES, and thus bypass liver and spleen filtration. Moreover, controlled release of the incorporated drug can be achieved for up to several weeks.26 The reduced volume of distribution of SLN is probably one of the reasons for the low amount of free drug present in tissues after SLN administration and could be related to slower distribution through biological barriers and/or the slow release from the SLN in the targeted organs.4 In particular, lower concentrations of free drug were observed in the heart after SLN than after free drug administration. Therefore, SLN might be useful to reduce the toxicity and to increase the clinical efficacy of anticancer drugs. Higher drug concentrations were detected in the brain in all of the studies when the drugs were encapsulated and delivered in SLN. Moreover, the low intrinsic toxicity and biodegradability of lipids used in SLN is a valuable feature in brain tumour treatment. In the late 1990s, SLN were proposed for brain drug targetting application independently by two research groups.109! 114 They developed scientific research to study the pharmacokinetics of two anticancer agents, namely camptothecin and doxorubicin, after the administration of free drug solution and drugloaded SLN to rats, observing drug accumulation into the brain after both oral and i.v. administration when loaded into SLN. Following SLN i.v. administration, the maximum concentration (Cmax ) increased by 180% when compared to the Cmax of the drug solution. The area under the curve (AUC)/dose and the MRT of SLN were 10.4- and 4-fold higher, respectively.58 In another independent study, stearic acid labelled SLN was found in rat CSF 20 min after i.v. administration. 30 min after administration of stealth SLN, the same doxorubicin concentration (around 10 "g/g of tissue) was found in the brain, heart, liver, lungs and spleen. However, after 24 hours brain doxorubicin concentrations remained higher than those observed in heart, liver, lungs and spleen. In recent years, various mechanisms for nanoparticle mediated drug uptake by the brain have been outlined. These include the enhancement of drug carrier retention in the brain blood capillaries by adsorption on to the capillary walls, resulting in a high concentration gradient across the blood brain barrier; the opening of tight junctions due to the presence of nanoparticles; the transcytosis of nanoparticles through the endothelium; solubilization of endothelial cell membrane lipids and membrane fluidization due to surfactant effects of polysorbates present in the drug carrier, or inhibition of efflux system of the blood brain barrier, especially P-glycoprotein.25 However, the mechanism for SLN uptake is not completely understood.

Lipid Nanomedicines for Anticancer Drug Therapy

6.4.1. Tumour Accumulation Several improvements in tumour drug accumulation have been observed when anticancer drugs are administered loaded to SLN. Elevated tumour etoposide concentrations were found when SLN were i.v. administered, compared to free drug to Dalton’s lymphoma tumour-bearing mice (67% increase 1 h post-injection, 30% increase 24 h postinjection).105 The accumulation of docetaxel in tumours was 1.7 to 2.4 times higher compared with the free drug at 6 h after intravenous injection to female nude mice bearing hepatoma.68 AUC values of DNA alkylating agent chlorambucil loaded SLN in tumour were significantly (p < 0.01) higher than the obtained after i.v. administration of the free drug to C57 BL/6 male mice subcutaneously inoculated with colon-38 tumour fragments from donor mice.67 The high accumulation of the anticancer drugs loaded in SLN could be attributed to following reasons; first, encapsulation of anticancer drugs into SLN resulted in higher accumulation in both the liver and the tumour. SLN can increase drug concentration in tumour via the previously explained EPR effect. Nanoparticles with a diameter below 200 nm can significantly accumulate in tumours 17

  19  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

REVIEW

Lipid Nanomedicines for Anticancer Drug Therapy

by this mechanism.118 Second, the internalization of SLN was further enhanced by the target of the SLN to specific carcinoma cells by modifying the nanoparticles with a ligand that can bind to specific receptor. As it has previously been mentioned in this review, the requirements of the antigen that will allow molecular targetting are the exclusive expression in cancer cells, to be an integral part of an essential cellular function of the cancer cells and not to easily mutate as the cancer cells proliferate.53 Docetaxel incorporated in galactosylated SLN showed a tumour increased accumulation in the treatment of hepatocellular carcinomas expressing the asialoglycoprotein receptor compared with unmodified SLN.68 On the other hand, a folate receptor-targeted SLN system significantly improved in vivo tumour growth inhibition and tumour bearing animal survival compared to non-targetted SLN.51 These findings will likely lead to the development of more targeted-SLN formulations designed for anticancer use. Significant improvements in tumour drug accumulation were observed when the etoposide formulations were injected intraperitoneally or subcutaneously rather than when intravenously administered.105 The slower and progressive penetration of the nanoparticles (and hence the loaded drug) from the peritoneum or the s.c. injection site into the tumour may result in more favourable patterns of drug distribution. The route of administration of a SLN formulation will be a key aspect to consider when designing animal or clinical studies using SLN for anticancer drug delivery. In another study, a 5-fold increase in AUCtumor compared to the free drug was observed after i.v. administration of PEGylated docetaxel-lipid nanocapsules to female BALB/c mice inoculated with C26 colon adenocarcinoma cells.52 Finally, a study was carried out with local injection of mitoxantrone SLN against breast cancer and its lymph node metastases.5 The drug concentration using SLN was much higher in local lymph nodes, and the drug concentration in other tissue was lower than that of mitoxantrone solution.

7. CONCLUDING REMARKS SLN-based anticancer drug delivery vehicles have been shown to be more efficient than conventional drug solutions, not only because the drug efficacy is improved while its toxicity is reduced, but also, because better pharmacokinetics and drug biodistribution profile were observed when the antitumour agent was encapsulated in these systems. Moreover, SLN have emerged as an effective alternative to liposomes, not only because of their better stability profile but also because they are easily scalable; since they are based on low cost biocompatible lipid excipients, their commercialisation will be easier compared to liposomes or polymeric nanoparticles. 18

Estella-Hermoso de Mendoza et al.et al. Mendoza

A certain number of lipid nanoparticulate systems have shown abilities to treat cancers that are usually refractory to cytotoxic drug treatment. Even though there is no SLNbased product for the treatment of cancer on the market so far, it can be expected that new modified types of SLN, such as NLC, LDC, stealth SLN, targeted-SLN, and SLN loaded with drug combinations, will be perfected and utilized to further improve the efficacies and side effect profiles of anticancer drugs. Consequently, the introduction of SLN to chemotherapy offers great promise for the management of cancerous diseases. Acknowledgments: We gratefully acknowledge support from the Spanish Ministry of Science and Innovation (SAF2007-61261, SAF2008-02251, PCT090100-2007-27) and Caja Navarra Foundation. Ander Estella-Hermoso de Mendoza is supported by the research grant from the Department of Education of the Basque Government (BF106.37).

References and Notes 1. R. B. Ewesuedo and M. J. Ratain, Oncologic therapeutics, edited by E. E. Vokes and H. M. Golomb (eds.). Springer, New York (2003), pp. 19–66. 2. C. H. Takimoto and E. Calvo, Cancer management: A multidisciplinary approach, 10th edn., edited by R. Pazdur, L. R. Coia, W. J. Hoskins, and L. D. Wagman (eds.). CMPMedica (2007). 3. G. Fontana, L. Maniscalco, D. Schillaci, G. Cavallaro, and G. Giammona, Solid lipid nanoparticles containing tamoxifen characterization and in vitro antitumoral activity. Drug Deliv. 12, 6 (2005). 4. A. Fundaro, R. Cavalli, A. Bargoni, D. Vighetto, G. P. Zara, and M. R. Gasco, Non-stealth and stealth solid lipid nanoparticles (SLN) carrying doxorubicin: pharmacokinetics and tissue distribution after i.v. administration to rats. Pharmacol. Res. 42, 4 (2000). 5. B. Lu, S. B. Xiong, H. Yang, X. D. Yin, and R. B. Chao, Solid lipid nanoparticles of mitoxantrone for local injection against breast cancer and its lymph node metastases. Eur. J. Pharm. Sci. 28, 1 (2006). 6. L. Serpe, M. G. Catalano, R. Cavalli, E. Ugazio, O. Bosco, R. Canaparo, E. Muntoni, R. Frairia, M. R. Gasco, M. Eandi, and G. P. Zara, Cytotoxicity of anticancer drugs incorporated in solid lipid nanoparticles on HT-29 colorectal cancer cell line. Eur. J. Pharm. Biopharm. 58, 3 (2004). 7. H. L. Wong, R. Bendayan, A. M. Rauth, Y. Li, and X. Y. Wu, Chemotherapy with anticancer drugs encapsulated in solid lipid nanoparticles. Adv. Drug Deliv. Rev. (2007). 8. W. J. Houlihan, M. Lohmeyer, P. Workman, and S. H. Cheon, Phospholipid antitumor agents. Med. Res. Rev. 15, 3 (1995). 9. F. Mollinedo, J. L. Fernandez-Luna, C. Gajate, B. Martin-Martin, A. Benito, R. Martinez-Dalmau, and M. Modolell, Selective induction of apoptosis in cancer cells by the ether lipid ET-18-OCH3 (Edelfosine): Molecular structure requirements, cellular uptake, and protection by Bcl-2 and Bcl-X(L). Cancer Res. 57, 7 (1997). 10. W. E. Berdel and S. Okamoto, Ether lipids in cancer chemotherapy. Keio J. Med. 39, 2 (1990). 11. A. Estella-Hermoso de Mendoza, M. A. Campanero, J. de la Iglesia-Vicente, C. Gajate, F. Mollinedo, and M. J. Blanco-Príeto, Anti-tumour alkyl ether lipid edelfosine: Tissue distribution and pharmacokinetic behaviour in healthy and tumour-bearing immune suppressed mice. Clin. Cancer Res. (2009), In press.

J. Biomed. Nanotechnol. 5, 1–21, 2009

  20  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso Mendoza et al. de Mendoza et al.

J. Biomed. Nanotechnol. 5, 1–21, 2009

34. M. Muchow, P. Maincent, and R. H. Muller, Lipid nanoparticles with a solid matrix (SLN, NLC, LDC) for oral drug delivery. Drug Dev. Ind. Pharm. 34, 12 (2008). 35. Z. R. Huang, S. C. Hua, Y. L. Yang, and J. Y. Fang, Development and evaluation of lipid nanoparticles for camptothecin delivery: A comparison of solid lipid nanoparticles, nanostructured lipid carriers, and lipid emulsion. Acta Pharmacol. Sin. 29, 9 (2008). 36. V. Jenning, K. Mader, and S. H. Gohla, Solid lipid nanoparticles (SLN) based on binary mixtures of liquid and solid lipids: a (1)HNMR study. Int. J. Pharm. 205, 1 (2000). 37. H. L. Wong, A. M. Rauth, R. Bendayan, J. L. Manias, M. Ramaswamy, Z. Liu, S. Z. Erhan, and X. Y. Wu, A new polymer-lipid hybrid nanoparticle system increases cytotoxicity of doxorubicin against multidrug-resistant human breast cancer cells. Pharm. Res. (2006). 38. R. H. Muller and C. M. Keck, Challenges and solutions for the delivery of biotech drugs–a review of drug nanocrystal technology and lipid nanoparticles. J. Biotechnol. 113, 1 (2004). 39. S. K. Jain, A. Chaurasiya, Y. Gupta, A. Jain, P. Dagur, B. Joshi, and V. M. Katoch, Development and characterization of 5-FU bearing ferritin appended solid lipid nanoparticles for tumour targeting. J. Microencapsul. 25, 5 (2008). 40. S. Markert, S. Lassmann, B. Gabriel, M. Klar, M. Werner, G. Gitsch, F. Kratz, and A. Hasenburg, Alpha-folate receptor expression in epithelial ovarian carcinoma and non-neoplastic ovarian tissue. Anticancer Res. 28, 6A (2008). 41. C. O. Evans, C. Yao, D. Laborde, and N. M. Oyesiku, Folate receptor expression in pituitary adenomas cellular and molecular analysis. Vitam. Horm. 79 (2008). 42. Y. Lu, E. Sega, and P. S. Low, Folate receptor-targeted immunotherapy: Induction of humoral and cellular immunity against haptendecorated cancer cells. Int. J. Cancer. 116, 5 (2005). 43. C. L. Denholt, P. R. Hansen, N. Pedersen, H. S. Poulsen, N. Gillings, and A. Kjaer, Identification of novel peptide ligands for the cancer-specific receptor mutation EFGRvIII using a mixturebased synthetic combinatorial library. Biopolymers 91, 3 (2009). 44. D. B. Cornelio, L. Meurer, R. Roesler, and G. Schwartsmann, Gastrin-releasing peptide receptor expression in cervical cancer. Oncology 73, 5 (2007). 45. K. Zhang, M. R. Aruva, N. Shanthly, C. A. Cardi, C. A. Patel, S. Rattan, G. Cesarone, E. Wickstrom, and M. L. Thakur, Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating peptide (PACAP) receptor specific peptide analogues for PET imaging of breast cancer: In vitro/in vivo evaluation. Regul. Pept. 144, 1 (2007). 46. M. G. Caruso, C. Messa, A. Orlando, B. D’Attoma, and M. Notarnicola, Early induction of LDL receptor gene expression by genistein in DLD-1 colon cancer cell line. Fitoterapia 79, 7 (2008). 47. C. Peterson, S. Vitols, M. Rudling, H. Blomgren, F. Edsmyr, and L. Skoog, Hypocholesterolemia in cancer patients may be caused by elevated LDL receptor activities in malignant cells. Med. Oncol. Tumor Pharmacother. 2, 3 (1985). 48. H. Yuan, J. Miao, Y. Z. Du, J. You, F. Q. Hu, and S. Zeng, Cellular uptake of solid lipid nanoparticles and cytotoxicity of encapsulated paclitaxel in A549 cancer cells. Int. J. Pharm. 348, 1 (2008). 49. X. G. Zhang, J. Miao, Y. Q. Dai, Y. Z. Du, H. Yuan, and F. Q. Hu, Reversal activity of nanostructured lipid carriers loading cytotoxic drug in multi-drug resistant cancer cells. Int. J. Pharm. 361, 1 (2008). 50. S. M. Stephenson, P. S. Low, and R. J. Lee, Folate receptormediated targeting of liposomal drugs to cancer cells. Methods Enzymol. 387 (2004). 51. P. J. Stevens, M. Sekido, and R. J. Lee, A folate receptor-targeted lipid nanoparticle formulation for a lipophilic paclitaxel prodrug. Pharm. Res. 21, 12 (2004).

REVIEW

12. F. Mollinedo, C. Gajate, S. Martin-Santamaria, and F. Gago, ET-18OCH3 (edelfosine): A selective antitumour lipid targeting apoptosis through intracellular activation of Fas/CD95 death receptor. Curr. Med. Chem. 11, 24 (2004). 13. C. Gajate and F. Mollinedo, The antitumor ether lipid ET-18OCH(3) induces apoptosis through translocation and capping of Fas/CD95 into membrane rafts in human leukemic cells. Blood. 98, 13 (2001). 14. J. Kotting, N. W. Marschner, W. Neumuller, C. Unger, and H. Eibl, Hexadecylphosphocholine and octadecyl-methyl-glycero3-phosphocholine: A comparison of hemolytic activity, serum binding and tissue distribution. Prog. Exp. Tumor Res. 34 (1992). 15. S. London, Intermittent oxaliplatin dosing prolongs time to treatment failure. Oncol. News Int. (2008). 16. K. Ruckmani, M. Sivakumar, and P. A. Ganeshkumar, Methotrexate loaded solid lipid nanoparticles (SLN) for effective treatment of carcinoma. J. Nanosci. Nanotechnol. 6, 9 (2006). 17. P. Speiser, Lipidnanopellets als tragersystem fur arzneimittel zur peroralen anwendung. Eur. Patent Appl. EP 0,167,825 (1986). 18. M. R. Gasco, Method for producing solid lipid microspheres having a narrow distribution. No. 5,250,236, (1993). 19. R. H. Müller and J. S. Lucks, Medication vehicles made of solid lipid particles (solid lipid nanospheres—SLN). No. 0605497, (1993). 20. J. Y. Fang, C. L. Fang, C. H. Liu, and Y. H. Su, Lipid nanoparticles as vehicles for topical psoralen delivery: Solid lipid nanoparticles (SLN) versus nanostructured lipid carriers (NLC). Eur. J. Pharm. Biopharm. 70, 2 (2008). 21. W. Mehnert and K. Mader, Solid lipid nanoparticles: production, characterization and applications. Adv. Drug Deliv. Rev. 47, 2 (2001). 22. R. H. Muller, K. Mader, and S. Gohla, Solid lipid nanoparticles (SLN) for controlled drug delivery—A review of the state of the art. Eur. J. Pharm. Biopharm. 50, 1 (2000). 23. S. S. Shidhaye, R. Vaidya, S. Sutar, A. Patwardhan, and V. J. Kadam, Solid lipid nanoparticles and nanostructured lipid carriers– innovative generations of solid lipid carriers. Curr. Drug Deliv. 5, 4 (2008). 24. C. Olbrich and R. H. Muller, Enzymatic degradation of SLN-effect of surfactant and surfactant mixtures. Int. J. Pharm. 180, 1 (1999). 25. Y. Chen, G. Dalwadi, and H. A. Benson, Drug delivery across the blood-brain barrier. Curr. Drug Deliv. 1, 4 (2004). 26. I. P. Kaur, R. Bhandari, S. Bhandari, and V. Kakkar, Potential of solid lipid nanoparticles in brain targeting. J. Control. Rel. (2008). 27. P. R. Lockman, J. M. Koziara, R. J. Mumper, and D. D. Allen, Nanoparticle surface charges alter blood-brain barrier integrity and permeability. J. Drug Target. 12, 9 (2004). 28. R. H. Muller and C. M. Keck, Drug delivery to the brain–realization by novel drug carriers. J. Nanosci. Nanotechnol. 4, 5 (2004). 29. H. Bunjes, F. Steiniger, and W. Richter, Visualizing the structure of triglyceride nanoparticles in different crystal modifications. Langmuir. 23, 7 (2007). 30. A. Estella-Hermoso de Mendoza, M. Rayo, F. Mollinedo, and M. J. Blanco-Prieto, Lipid nanoparticles for alkyl lysophospholipid edelfosine encapsulation: Development and in vitro characterization. Eur. J. Pharm. Biopharm. 68, 2 (2008). 31. E. B. Souto, W. Mehnert, and R. H. Muller, Polymorphic behaviour of Compritol888 ATO as bulk lipid and as SLN and NLC. J. Microencapsul. 23, 4 (2006). 32. K. Manjunath, J. S. Reddy, and V. Venkateswarlu, Solid lipid nanoparticles as drug delivery systems. Meth. Find. Exp. Clin. Pharmacol. 27, 2 (2005). 33. A. Zur Muhlen, C. Schwarz, and W. Mehnert, Solid lipid nanoparticles (SLN) for controlled drug delivery–drug release and release mechanism. Eur. J. Pharm. Biopharm. 45, 2 (1998).

Lipid Nanomedicines for Anticancer Drug Therapy

19

  21  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

REVIEW

Lipid Nanomedicines for Anticancer Drug Therapy 52. M. N. Khalid, P. Simard, D. Hoarau, A. Dragomir, and J. C. Leroux, Long circulating poly(ethylene glycol)-decorated lipid nanocapsules deliver docetaxel to solid tumors. Pharm. Res. 23, 4 (2006). 53. R. Abou-Jawde, T. Choueiri, C. Alemany, and T. Mekhail, An overview of targeted treatments in cancer. Clin. Ther. 25, 8 (2003). 54. B. Stella, S. Arpicco, M. T. Peracchia, D. Desmaele, J. Hoebeke, M. Renoir, J. D’Angelo, L. Cattel, and P. Couvreur, Design of folic acid-conjugated nanoparticles for drug targeting. J. Pharm. Sci. 89, 11 (2000). 55. A. Beduneau, P. Saulnier, F. Hindre, A. Clavreul, J. C. Leroux, and J. P. Benoit, Design of targeted lipid nanocapsules by conjugation of whole antibodies and antibody Fab’ fragments. Biomaterials 28, 33 (2007). 56. Y. Gupta, A. Jain, and S. K. Jain, Transferrin-conjugated solid lipid nanoparticles for enhanced delivery of quinine dihydrochloride to the brain. J. Pharm. Pharmacol. 59, 7 (2007). 57. S. J. Lim and C. K. Kim, Formulation parameters determining the physicochemical characteristics of solid lipid nanoparticles loaded with all-trans retinoic acid. Int. J. Pharm. 243, 1 (2002). 58. S. C. Yang, L. F. Lu, Y. Cai, J. B. Zhu, B. W. Liang, and C. Z. Yang, Body distribution in mice of intravenously injected camptothecin solid lipid nanoparticles and targeting effect on brain. J. Control. Rel. 59, 3 (1999). 59. L. H. Reddy, J. S. Adhikari, B. S. Dwarakanath, R. K. Sharma, and R. R. Murthy, Tumoricidal effects of etoposide incorporated into solid lipid nanoparticles after intraperitoneal administration in Dalton’s lymphoma bearing mice. AAPS J. 8, 2 (2006). 60. L. H. Reddy, R. K. Sharma, K. Chuttani, A. K. Mishra, and R. R. Murthy, Etoposide-incorporated tripalmitin nanoparticles with different surface charge: formulation, characterization, radiolabeling, and biodistribution studies. AAPS J. 6, 3 (2004). 61. M. K. Lee, S. J. Lim, and C. K. Kim, Preparation, characterization and in vitro cytotoxicity of paclitaxel-loaded sterically stabilized solid lipid nanoparticles. Biomaterials 28, 12 (2007). 62. J. You, F. Wan, F. de Cui, Y. Sun, Y. Z. Du, and F. Q. Hu, Preparation and characteristic of vinorelbine bitartrate-loaded solid lipid nanoparticles. Int. J. Pharm. 343, 1 (2007). 63. Y. Hattori and Y. Maitani, Folate-linked lipid-based nanoparticle for targeted gene delivery. Curr. Drug Deliv. 2, 3 (2005). 64. K. Tabatt, C. Kneuer, M. Sameti, C. Olbrich, R. H. Muller, C. M. Lehr, and U. Bakowsky, Transfection with different colloidal systems: Comparison of solid lipid nanoparticles and liposomes. J. Control. Rel. 97, 2 (2004). 65. K. Tabatt, M. Sameti, C. Olbrich, R. H. Muller, and C. M. Lehr, Effect of cationic lipid and matrix lipid composition on solid lipid nanoparticle-mediated gene transfer. Eur. J. Pharm. Biopharm. 57, 2 (2004). 66. J. Tian, X. Pang, K. Yu, L. Liu, and J. Zhou, Preparation, characterization and in vivo distribution of solid lipid nanoparticles loaded with cisplatin. Pharmazie 63, 8 (2008). 67. P. Sharma, S. Ganta, W. A. Denny, and S. Garg, Formulation and pharmacokinetics of lipid nanoparticles of a chemically sensitive nitrogen mustard derivative: Chlorambucil. Int. J. Pharm. (2008). 68. Z. Xu, L. Chen, W. Gu, Y. Gao, L. Lin, Z. Zhang, Y. Xi, and Y. Li, The performance of docetaxel-loaded solid lipid nanoparticles targeted to hepatocellular carcinoma. Biomaterials 30, 2 (2009). 69. A. Miglietta, R. Cavalli, C. Bocca, L. Gabriel, and M. R. Gasco, Cellular uptake and cytotoxicity of solid lipid nanospheres (SLN) incorporating doxorubicin or paclitaxel. Int. J. Pharm. 210, 1 (2000). 70. G. P. Zara, R. Cavalli, A. Bargoni, A. Fundaro, D. Vighetto, and M. R. Gasco, Intravenous administration to rabbits of non-stealth and stealth doxorubicin-loaded solid lipid nanoparticles at increasing concentrations of stealth agent: pharmacokinetics and distribution of doxorubicin in brain and other tissues. J. Drug Target. 10, 4 (2002).

20

Mendoza Estella-Hermoso de Mendoza et al.et al. 71. G. P. Zara, A. Bargoni, R. Cavalli, A. Fundaro, D. Vighetto, and M. R. Gasco, Pharmacokinetics and tissue distribution of idarubicin-loaded solid lipid nanoparticles after duodenal administration to rats. J. Pharm. Sci. 91, 5 (2002). 72. L. Serpe, M. Guido, R. Canaparo, E. Muntoni, R. Cavalli, P. Panzanelli, C. Della Pepal, A. Bargoni, A. Mauro, M. R. Gasco, M. Eandi, and G. P. Zara, Intracellular accumulation and cytotoxicity of doxorubicin with different pharmaceutical formulations in human cancer cell lines. J. Nanosci. Nanotechnol. 6, 9 (2006). 73. M. Trotta, F. Debernardi, and O. Caputo, Preparation of solid lipid nanoparticles by a solvent emulsification-diffusion technique. Int. J. Pharm. 257, 1 (2003). 74. A. Del Pozo-Rodriguez, M. A. Solinis, A. R. Gascon, and J. L. Pedraz, Short- and long-term stability study of lyophilized solid lipid nanoparticles for gene therapy. Eur. J. Pharm. Biopharm. (2008). 75. R. Paliwal, S. Rai, B. Vaidya, K. Khatri, A. K. Goyal, N. Mishra, A. Mehta, and S. P. Vyas, Effect of lipid core material on characteristics of solid lipid nanoparticles designed for oral lymphatic delivery. Nanomedicine: Nanotechnology, Biology and Medicine. Corrected Proof (2008), In Press. 76. R. Cortesi, E. Esposito, G. Luca, and C. Nastruzzi, Production of lipospheres as carriers for bioactive compounds. Biomaterials 23, 11 (2002). 77. M. Uner, Preparation, characterization and physico-chemical properties of solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC): their benefits as colloidal drug carrier systems. Pharmazie 61, 5 (2006). 78. Y. Wang, Y. Deng, S. Mao, S. Jin, J. Wang, and D. Bi, Characterization and body distribution of beta-elemene solid lipid nanoparticles (SLN). Drug Dev. Ind. Pharm. 31, 8 (2005). 79. E. Zimmermann, R. H. Muller, and K. Mader, Influence of different parameters on reconstitution of lyophilized SLN. Int. J. Pharm. 196, 2 (2000). 80. W. N. Charman and V. J. Stella, Lymphatic Transport of Drugs, CRC Press (1992). 81. J. Kreuter, Physicochemical characterization of polyacrylic nanoparticles. Int. J. Pharm. 14, 1 (1983). 82. M. Nidhin, R. Indumathy, K. J. Sreeram, and B. U. Nair, Synthesis of iron oxide nanoparticles of narrow size distribution on polysaccharide templates. Bull. Mater. Sci. 31, 1 (2008). 83. P. Ahlin, J. Kristl, A. Kristl, and F. Vrecer, Investigation of polymeric nanoparticles as carriers of enalaprilat for oral administration. Int. J. Pharm. 239, 1 (2002). 84. J. Stecova, W. Mehnert, T. Blaschke, B. Kleuser, R. Sivaramakrishnan, C. C. Zouboulis, H. Seltmann, H. C. Korting, K. D. Kramer, and M. Schafer-Korting, Cyproterone acetate loading to lipid nanoparticles for topical acne treatment: particle characterisation and skin uptake. Pharm. Res. 24, 5 (2007). 85. J. Zhang, Y. Fan, and E. Smith, Experimental design for the optimization of lipid nanoparticles. J. Pharm. Sci. (2008). 86. N. Anton, J. P. Benoit, and P. Saulnier, Design and production of nanoparticles formulated from nano-emulsion templates-a review. J. Control. Rel. 128, 3 (2008). 87. C. Freitas and R. H. Muller, Effect of light and temperature on zeta potential and physical stability in solid lipid nanoparticles (SLN) dispersion. Int. J. Pharm. 168 (1998). 88. N. Gadegaard, Atomic force microscopy in biology: technology and techniques. Biotech. Histochem. 81, 2 (2006). 89. H. Zhang, W. Cui, J. Bei, and S. Wang, Preparation of poly(lactideco-glycolide-co-caprolactone) nanoparticles and their degradation behaviour in aqueous solution. Polym. Degrad. Stab. 91, 9 (2006). 90. A. Radomska-Soukharev, Stability of lipid excipients in solid lipid nanoparticles. Adv. Drug Deliv. Rev. 59, 6 (2007). 91. T. Kramer, D. M. Kremer, M. J. Pikal, W. J. Petre, E. Y. Shalaev, and L. A. Gatlin, A procedure to optimize scale-up for the primary drying phase of lyophilization. J. Pharm. Sci. 98, 1 (2009).

J. Biomed. Nanotechnol. 5, 1–21, 2009

  22  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

Estella-Hermoso Mendoza et al. de Mendoza et al.

107. A. Bargoni, R. Cavalli, O. Caputo, A. Fundaro, M. R. Gasco, and G. P. Zara, Solid lipid nanoparticles in lymph and plasma after duodenal administration to rats. Pharm. Res. 15, 5 (1998). 108. N. L. Trevaskis, W. N. Charman, and C. J. Porter, Lipid-based delivery systems and intestinal lymphatic drug transport: a mechanistic update. Adv. Drug Deliv. Rev. 60, 6 (2008). 109. G. P. Zara, R. Cavalli, A. Fundaro, A. Bargoni, O. Caputo, and M. R. Gasco, Pharmacokinetics of doxorubicin incorporated in solid lipid nanospheres (SLN). Pharmacol. Res. 40, 3 (1999). 110. H. Yuan, J. Chen, Y. Z. Du, F. Q. Hu, S. Zeng, and H. L. Zhao, Studies on oral absorption of stearic acid SLN by a novel fluorometric method. Colloids Surf. B Biointerfaces 58, 2 (2007). 111. L. Hu, X. Tang, and F. Cui, Solid lipid nanoparticles (SLNs) to improve oral bioavailability of poorly soluble drugs. J. Pharm. Pharmacol. 56, 12 (2004). 112. S. Peltier, J. M. Oger, F. Lagarce, W. Couet, and J. P. Benoit, Enhanced oral paclitaxel bioavailability after administration of paclitaxel-loaded lipid nanocapsules. Pharm. Res. 23, 6 (2006). 113. H. Chen, X. Chang, D. Du, W. Liu, J. Liu, T. Weng, Y. Yang, H. Xu, and X. Yang, Podophyllotoxin-loaded solid lipid nanoparticles for epidermal targeting. J. Control. Rel. 110, 2 (2006). 114. S. Yang, J. Zhu, Y. Lu, B. Liang, and C. Yang, Body distribution of camptothecin solid lipid nanoparticles after oral administration. Pharm. Res. 16, 5 (1999). 115. D. E. Owens, 3rd and N. A. Peppas, Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm. 307, 1 (2006). 116. C. E. Soma, C. Dubernet, G. Barratt, S. Benita, and P. Couvreur, Investigation of the role of macrophages on the cytotoxicity of doxorubicin and doxorubicin-loaded nanoparticles on M5076 cells in vitro. J. Control. Rel. 68, 2 (2000). 117. J. Gualbert, P. Shahgaldian, and A. W. Coleman, Interactions of amphiphilic calix[4]arene-based Solid Lipid Nanoparticles with bovine serum albumin. Int. J. Pharm. 257, 1 (2003). 118. N. Iodoshima, C. Udagawa, T. Ando, H. Fukuyasu, T. Watanabe, and S. Nakabayashi, Lipid nanoparticles for delivering antitumor drugs. Int. J. Pharm. 146 (1997). 119. J. X. Wang, X. Sun, and Z. R. Zhang, Enhanced brain targeting by synthesis of 3! ,5! -dioctanoyl-5-fluoro-2! -deoxyuridine and incorporation into solid lipid nanoparticles. Eur. J. Pharm. Biopharm. 54, 3 (2002). 120. L. H. Reddy, K. Vivek, N. Bakshi, and R. S. Murthy, Tamoxifen citrate loaded solid lipid nanoparticles (SLN): preparation, characterization, in vitro drug release, and pharmacokinetic evaluation. Pharm. Dev. Technol. 11, 2 (2006).

REVIEW

92. P. Yadava, M. Gibbs, C. Castro, and J. A. Hughes, Effect of lyophilization and freeze-thawing on the stability of siRNAliposome complexes. AAPS Pharm. Sci. Tech. 9, 2 (2008). 93. Y. Bensouda and A. Laatiris, The lyophilization of dispersed systems: influence of freezing process, freezing time, freezing temperature and RBCs concentration on RBCs hemolysis. Drug Dev. Ind. Pharm. 32, 8 (2006). 94. R. Seetharam, Y. Wada, S. Ramachandran, H. Hess, and P. Satir, Long-term storage of bionanodevices by freezing and lyophilization. Lab. Chip. 6, 9 (2006). 95. R. Cavalli, O. Caputo, and M. R. Gasco, Preparation and characterization of solid lipid nanospheres containing paclitaxel. Eur. J. Pharm. Sci. 10, 4 (2000). 96. C. Schwarz and W. Mehnert, Freeze-drying of drug-free and drugloaded solid lipid nanoparticles (SLN). Int. J. Pharm. 157, 2 (1997). 97. R. H. Müller, S. Maassen, C. Schwarz, and W. Mehnert, Solid lipid nanoparticles (SLN) as potential carrier for human use: interaction with human granulocytes. J. Control. Rel. 47, 3 (1997). 98. N. Scholer, H. Hahn, R. H. Muller, and O. Liesenfeld, Effect of lipid matrix and size of solid lipid nanoparticles (SLN) on the viability and cytokine production of macrophages. Int. J. Pharm. 231, 2 (2002). 99. H. Weyhers, S. Ehlers, H. Hahn, E. B. Souto, and R. H. Muller, Solid lipid nanoparticles (SLN)–effects of lipid composition on in vitro degradation and in vivo toxicity. Pharmazie 61, 6 (2006). 100. M. D. Joshi and R. H. Muller, Lipid nanoparticles for parenteral delivery of actives. Eur. J. Pharm. Biopharm. 71, 2 (2009). 101. P. K. Dudeja, K. M. Anderson, J. S. Harris, L. Buckingham, and J. S. Coon, Reversal of multidrug resistance phenotype by surfactants: relationship to membrane lipid fluidity. Arch. Biochem. Biophys. 319, 1 (1995). 102. D. M. Woodcock, M. E. Linsenmeyer, G. Chojnowski, A. B. Kriegler, V. Nink, L. K. Webster, and W. H. Sawyer, Reversal of multidrug resistance by surfactants. Br. J. Cancer. 66, 1 (1992). 103. E. Allard, C. Passirani, E. Garcion, P. Pigeon, A. Vessieres, G. Jaouen, and J. P. Benoit, Lipid nanocapsules loaded with an organometallic tamoxifen derivative as a novel drug-carrier system for experimental malignant gliomas. J. Control. Rel. 130, 2 (2008). 104. D. B. Chen, T. Z. Yang, W. L. Lu, and Q. Zhang, In vitro and in vivo study of two types of long-circulating solid lipid nanoparticles containing paclitaxel. Chem. Pharm. Bull. (Tokyo). 49, 11 (2001). 105. L. H. Reddy, R. K. Sharma, K. Chuttanib, A. K. Mishrab, and R. S. R. Murth, Influence of administration route on tumor uptake and biodistribution of etoposide loaded solid lipid nanoparticles in Dalton’s lymphoma tumor bearing mice. J. Control. Rel. 105 (2005). 106. N. Scholer, C. Olbrich, K. Tabatt, R. H. Muller, H. Hahn, and O. Liesenfeld, Surfactant, but not the size of solid lipid nanoparticles (SLN) influences viability and cytokine production of macrophages. Int. J. Pharm. 221, 1 (2001).

Lipid Nanomedicines for Anticancer Drug Therapy

  Received: xx Xxxx xxxx. Accepted: xx Xxxx xxxx.

J. Biomed. Nanotechnol. 5, 1–21, 2009

21

  23  

 

 

 

               

 

     

Hypothesis  and  Objectives   -­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐-­‐   Hipótesis  y  Objetivos      

25  

 

 

 

  Hypothesis  and  Objectives    

HYPOTHESIS  AND  OBJECTIVES   The   synthetic   analogues,   ether   phospholipids,   also   known   as   alkyl-­‐ lysophospholipids   (ALP),   among   which   1-­‐O-­‐octadecyl-­‐2-­‐O-­‐methyl-­‐rac-­‐glycero-­‐3-­‐ phosphocholine   (ET-­‐18-­‐OCH3,   edelfosine)   is   the   prototypical   compound,   constitute  a  promising  class  of  oral  anti-­‐tumor  substances  which  do  not  have  DNA   as  their  target,  but  rather  act  at  the  level  of  the  membrane.  However,  due  to  their   chemical   structure,   these   molecules   present   a   dose   depending   hemolysis   and   gastrointestinal  toxicity  when  they  are  administered  at  high  doses,  as  well  as  low   oral  bioavailability.   On   the   other   hand,   lipid   nanoparticles   (LN)   have   arisen   as   an   interesting   proposal   for   the   oral   administration   of   drugs,   since   due   to   their   characteristics   they   facilitate   the   solubilization   of   highly   lipophilic   substances   in   addition   to   favoring   the   absorption   of   the   drugs   in   the   intestine.   In   fact,   these   particles   have   been   used   to   increase   or   improve   the   oral   bioavailability   of   numerous   active   principles,   since   due   to   their   small   size   and   composition   LN   can   penetrate   into   the   spaces  between  the  villi  of  the  intestinal  mucosa  and  thus  reach  the  blood  stream.   The   use   of   these   nanotransporters   for   the   transport   of   anti-­‐tumor   agents   additionally  offers  an  important  advantage:  their  tumor  specificity  due  to  the  EPR   effect   (“enhanced   permeability   and   retention   effect”)   and   therefore   their   lower   toxicity.   On  the  basis  of  the  foregoing,  the  initial  hypothesis  of  this  work  is:     The  use  of  LN  of  edelfosine  may  produce  a  highly  significant  increase  in  the  anti-­‐ tumor  efficacy  of  this  compound,  both  because  it  may  propitiate  absorption  in  the   intestine,   and   because   it   would   reduce   the   dose   of   active   principle   needed,   since   high  daily  doses  would  not  be  necessary  to  maintain  significant  drug  levels  in  the   blood,  which  would  also  lessen  any  possible  toxic  effects.        

27  

  Hypothesis  and  Objectives    

  In   short,   it   is   expected   that   oral   administration   of   edelfosine   inside   these   nanotransporters  will  result  in  an  increase  in  the  bioavailability  of  the  drug,  and  a   reduction  in  both  the  required  dose  and  the  possible  adverse  reactions  associated   with  this  drug.     The   general   aim   of   this   study   is   to   design,   develop   and   evaluate   lipid   nanotransporters  containing  edelfosine  both  in  vitro  and  in  vivo.      

  More  specifically,  the  partial  objectives  are  as  follows:   1. Determination   of   the   pharmacokinetic   profile   of   free   edelfosine   and   its   biodistribution   in   mantle   cell   lymphoma-­‐bearing   and   non   mantle   cell   lymphoma-­‐bearing  animal  models.   2. Development,   formulation   and   physico-­‐chemical   characterization   of   lipid   drug  nanotransporters.   3. Determination  of  the  pharmacokinetic  profile  of  edelfosine  when  vectorized   into  LN  and  its  biodistribution  in  animal  models.   4. Evaluation   of   the   (therapeutic)   efficacy   of   the   selected   nanomedicines   in   animal  models  of  cancer  (mantle  cell  lymphoma  and  glioma).  

28  

  Hipótesis  y  Objetivos    

HIPOTESIS  Y  OBJETIVOS   Los   análogos   sintéticos   éter   fosfolípidos,   o   también   conocidos   como   alquil-­‐ lisofosfolípidos   entre   los   que   el   1-­‐O-­‐octadecil-­‐2-­‐O-­‐metil-­‐rac-­‐glicero-­‐3-­‐fosfocolina   (ET-­‐18-­‐OCH3,   edelfosina)   es   su   compuesto   prototípico,   constituyen   una   prometedora  clase  de  substancias  antitumorales  orales  que  no  tienen  al  ADN  como   diana,   sino   que   actúan   al   nivel   de   la   membrana.   Sin   embargo,   y   debido   a   su   naturaleza   química,   estas   moléculas   poseen   una   toxicidad   hemolítica   dosis   dependiente,   además   de   toxicidad   en   el   tracto   gastrointestinal   cuando   se   administran  a  dosis  elevadas  y  baja  biodisponibilidad  oral.   Por   otro   lado,   las   nanopartículas   lipídicas   (NL)   han   surgido   como   una   propuesta   prometedora   para   la   administración   oral   de   fármacos,   ya   que   por   sus   características   facilitan   la   solubilización   de   sustancias   muy   lipófilas   además   de   favorecer  la  absorción  de  los  fármacos  a  nivel  intestinal.  De  hecho,  estas  partículas   han   sido   utilizadas   para   aumentar   o   mejorar   la   biodisponibilidad   oral   de   numerosos   principios   activos   ya   que   debido   a   su   pequeño   tamaño   y   a   su   composición,   las   LN   pueden   penetrar   en   los   espacios   entre   las   villi   de   la   mucosa   intestinal   y   llegar   al   torrente   circulatorio.   El   uso   de   estos   nanotransportadores   para   el   transporte   de   agentes   antitumorales   ofrece   además   una   importante   ventaja:  su  especificidad  tumoral  debido  a  un  efecto  realzado  de  la  permeabilidad  y   la  retención  en  la  zona  peritumoral  y  por  tanto,  una  menor  toxicidad.   Teniendo  en  cuenta  todo  lo  anterior,  la  hipótesis  de  partida  de  este  trabajo  es  la   siguiente:   El  uso  de  nanopartículas  lipídicas  de  edelfosina  podría  suponer  un  incremento   muy   significativo   de   la   eficacia   antitumoral   de   este   compuesto,   tanto   por   la   posibilidad   de   aumentar   la   absorción   a   nivel   intestinal   como   por   la   de   disminuir   la   dosis   de   principio   activo,   no   requiriéndose   por   tanto   una   dosis   diaria   elevada   para   mantener   niveles   de   compuesto   farmacológicamente   significativos   en   sangre,   lo   que  a  su  vez  disminuirá  los  posibles  efectos  tóxicos.     29  

  Hipótesis  y  Objetivos    

  En   definitiva,   se   espera   que   la   administración   por   vía   oral   de   la   edelfosina   vehiculizada   en   estos   nanotrasportadores   se   traduzca   en   un   aumento   de   la   biodisponibilidad  del  fármaco,  así  como  en  la  disminución  de  la  dosis  administrada   y  posibles  reacciones  adversas  asociadas  a  éste  fármaco.     El  objetivo  general  de  este  trabajo  es  el  diseño,  desarrollo  y  evaluación  in  vitro   e  in  vivo  de  nanotransportadores  lipídicos  conteniendo  edelfosina.     Los  objetivos  parciales  son  específicamente:   1. Determinación   del   perfil   farmacocinético   de   la   edelfosina   y   su   biodistribución  en  modelos  animales  tanto  con  linfoma  de  manto  como  sin   él.   2. Desarrollo,  

formulación  

y  

caracterización  

físico-­‐química  

de  

nanotransportadores  lipídicos  del  fármaco.   3. Determinación   del   perfil   farmacocinético   de   la   edelfosina   vectorizada   en   nanopartículas  lipídicas  (NL)  y  su  biodistribución  en  modelos  animales.   4. Evaluación  de  la  eficacia  (terapéutica)  de  los  nanomedicinas  seleccionadas   en  modelos  animales  de  cáncer  (linfoma  de  manto  y  glioma).    

30  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

 

Chapter  1   Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution   and  pharmacokinetic  behavior  in  healthy  and  tumor-­‐ bearing  immunosuppressed  mice   Ander  Estella-­‐Hermoso  de  Mendoza1,  Miguel  A.  Campanero2,  Janis  de  la  Iglesia-­‐ Vicente3,  Consuelo  Gajate3,4,  Faustino  Mollinedo3,  María  J.  Blanco-­‐Prieto1     1Departamento  de  Farmacia  y  Tecnología  Farmacéutica,  Facultad  de  Farmacia,  

University  of  Navarra,  E-­‐31008,  Spain   2Servicio  de  Farmacología  Clínica,  Clínica  Universitaria,  E-­‐31080  Pamplona,  Spain   3Centro  de  Investigación  del  Cáncer,  Instituto  de  Biología  Molecular  y  Celular  del  

Cáncer,  CSIC-­‐Universidad  de  Salamanca,  Campus  Miguel  de  Unamuno,  E-­‐37007   Salamanca,  Spain   4Unidad  de  Investigación,  Hospital  Universitario  de  Salamanca,  Campus  Miguel  de  

Unamuno,  E-­‐37007  Salamanca,  Spain     Running  Title:  Biodistribution  and  pharmacokinetics  of  edelfosine   Key   Words:   edelfosine,   biodistribution,   mantle   cell   lymphoma,   bioavailability,   pharmacokinetics   Corresponding   author:   Dr.   María   J.   Blanco-­‐Prieto,   Department   of   Pharmacy   and   Pharmaceutical   Technology,   School   of   Pharmacy,   University   of   Navarra,   C/Irunlarrea   1,   E-­‐31080   Pamplona,   Spain,   Office   phone:   +   34   948   425   600   ext.   6519,  Fax:  +  34  948  425  649,  e-­‐mail:  [email protected]    

 

Clinical  Cancer  Research,  2009.  15(3):  p.  858-­‐64

31  

 

 

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

 

Abstract     Purpose:   The   present   study   investigates   and   compares   the   dose-­‐dependent   pharmacokinetics   and   oral   bioavailability   of   edelfosine   in   healthy,   immune   deficient   and   tumour-­‐bearing   immune   suppressed   mouse   animal   models,   as   well   as   edelfosine   uptake   and   apoptotic   activity   in   the   Z-­‐138   mantle   cell   lymphoma   (MCL)  cell  line.       Experimental   design:   Biodistribution   study   of   edelfosine   was   performed   in   both  BALB/c  and  severe  combined  immune  deficiency  (SCID)  mice,  and  then  the  in   vivo   behaviour   of   the   drug   after   intravenous   and   oral   administration   was   monitored.       Results:   We   found   that   edelfosine   is   incorporated   and   induces   apoptosis   in   the   Z-­‐138   human   mantle   cell   lymphoma   cell   line,   whereas   normal   resting   peripheral   blood   human   lymphocytes   were   not   affected.   In   vivo   biodistribution   studies   revealed   that   accumulation   of   edelfosine   in   the   tumour   of   a   MCL-­‐bearing   mouse  animal  model  was  considerably  higher  (p  <  0.01)  than  in  the  other  organs   analysed.   Besides,   no   statistical   differences   were   observed   between   the   pharmacokinetic  parameters  of  BALB/c  and  SCID  mice.  Edelfosine  presented  slow   elimination  and  high  distribution  to  tissues.  Bioavailability  for  a  single  oral  dose  of   edelfosine   was   less   than   10   %,   but   a   multiple   dose   oral   administration   increased   this  value  up  to  64  %.       Conclusion:   Our   results   show   that   edelfosine   is   widely   scattered   across   different   organs,   but   that   it   is   preferentially   internalised   by   the   tumour   both   in   vitro  and  in  vivo.        

33  

  Introduction.  Lipid  nanomedicines  for  anticancer  drug  therapy    

34  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

1. Introduction   Edelfosine   is   the   prototype   molecule   of   a   promising   family   of   anti-­‐tumour   compounds  collectively  known  as  synthetic  alkyl-­‐lysophospholipids,  which  include   also  clinically  relevant  drugs  such  as  miltefosine  and  perifosine,  and  which  can  be   administered  orally  [1-­‐4].  This  class  of  synthetic  anti-­‐cancer  agents  acts  at  the  level   of  the  cell  membrane,  unlike  most  currently  available  chemotherapeutic  drugs  that   target   the   nuclear   DNA,   and   induces   selective   apoptosis   in   malignant   cells,   sparing   normal   cells   [5-­‐7].   Although   the   precise   mechanism   of   action   is   not   yet   fully   elucidated,  edelfosine-­‐induced  apoptosis  is  mediated  by  Fas/CD95  death  receptor   [8-­‐10],   JNK   activation   [11,   12],   endoplasmic   reticulum   stress   [13]   and   mitochondria   [14].   Edelfosine   has   been   applied   as   a   purging   agent   in   autologous   bone   marrow   transplantation   [15],   and   recent   data   suggest   a   putative   clinical   relevance   for   this   agent   in   cancer   [7].   However,   there   is   very   little   information   regarding  the  disposition  and  oral  bioavailability  of  edelfosine  in  vivo.   The   kinetic   behaviour   of   radioactive   edelfosine   in   rats   and   mice   after   intravenous   administration   has   been   previously   investigated.   Arnold   et   al.   [16]   administered  15  daily  injections  of  an  amount  of  radioactive  edelfosine  of  1  ·  106   cpm   (counts   per   minute)   intravenously   to   methylcholantrene-­‐induced   fibrosarcoma-­‐bearing   mice.   Results   showed   that   1   day   after   the   last   injection   an   overall   activity   of   only   2   ·   106   cpm   was   recovered.   This   indicated   that   edelfosine   did  not  accumulate  in  the  organism.  However,  measurement  of  total  radioactivity   does   not   reflect   the   pharmacokinetic   behaviour   or   the   tissue   distribution   of   the   parent  drug  edelfosine,  since  the  radiolabel  may  be  included  in  some  metabolites.   Kötting   et   al.   studied   biodistribution   in   rats   after   oral   administration   of   edelfosine   [17].   Quantification   was   performed   with   high   performance   thin-­‐layer   chromatography   (HPTLC).     After   24   h,   no   significant   levels   of   edelfosine   were   detected   either   in   faeces   or   urine.   This   information   led   them   to   conclude   that   approximately  96  %  of  the  drug  was  absorbed  in  the  first  24  h  [17].         As   yet   no   precise   pharmacokinetic   parameters   have   been   determined   for   free   edelfosine.   Characterisation   of   the   pharmacokinetics   of   edelfosine   is   crucial   to  

35  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

understand   the   in   vivo   concentration-­‐effect   and   concentration-­‐toxicity   relationships   and   to   choose   dosing   regimens.   Furthermore,   knowledge   of   the   in   vivo   concentration   range,   oral   bioavailability   and   pharmacokinetics   of   free   edelfosine   is   crucial   to   evaluate   the   effects   of   edelfosine   delivery   systems   on   the   pharmacokinetic  behaviour  of  this  anti-­‐neoplastic  agent.   In   order   to   determine   levels   of   edelfosine   either   in   plasma,   tissue   or   tumour,   sensitive   and   selective   techniques   must   be   used   to   detect   only   the   compound   of   interest   and   not   others,   like   metabolites   or   very   similar   molecules.   We   have   recently   developed   a   simple   and   highly   selective   and   sensitive   HPLC-­‐MS   technique   with   a   quantitation   limit   of   0.3   ng,   which   avoids   the   use   of   radiolabeled   compounds   [18].   With   this   technique   it   is   possible   to   quantify   edelfosine   concentrations   either   in   plasma,   tissues   or   tumour   accurately   and   sensitively   in   order  to  determine  pharmacokinetic  parameters.   The   present   study   investigates   and   compares   the   dose-­‐dependent   pharmacokinetics   and   oral   bioavailability   of   edelfosine   in   healthy,   immune   deficient  and  tumour-­‐bearing  immune  suppressed  mouse  animal  models.       Severe  combined  immune  deficiency  (SCID)  mice  are  routinely  used  as  hosts  for   malignant   cells   and   for   in   vivo   testing   of   new   anti-­‐tumour   agents.   SCID   mice   are   characterised   by   the   complete   inability   of   the   adaptive   immune   system   to   mount   an   appropriate   immune   response,   due   to   absence   of   functional   lymphocytes   as   a   result  of  defects  in  T  and  B  cell  development.  On  these  grounds,  use  of  SCID  mice   permits  the  long-­‐term  engraftment  of  human  tumour  cells.  Because  most  of  the  in   vivo   anti-­‐tumour   drug   testing   assays   are   carried   out   with   SCID   mice,   accomplishment   of   the   pharmacokinetics   in   these   immune   compromised   animals   is  imperative,  and  so  we  studied  the  tissue  and  tumour  drug  distribution  in  these   immune  deficient  mice.  In  addition,  in  vitro  experiments  with  the  Z-­‐138  mantle  cell   lymphoma  (MCL)  cell  line,  used  for  xenograft  assays,  were  performed  in  order  to   determine  both  edelfosine  uptake  and  apoptotic  activity.      

36  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

2. Materials  and  Methods   2.1.

Chemicals.    

Edelfosine   was   from   INKEYSA   (Barcelona,   Spain).   Platelet   Activating   Factor   (PAF)  and  PBS  [10  mM  phosphate,  0.9  %  NaCl]  were  obtained  from  Sigma-­‐Aldrich   (Madrid,  Spain).  Formic  acid  99  %  was  purchased  from  Fluka  (Madrid,  Spain)  and   methanol  was  obtained  from  Merck  (Barcelona,  Spain).  All  solvents  employed  for   the   analysis   were   of   analytical   grade.   RPMI-­‐1640   cell   culture   medium,   heat-­‐ inactivated   fetal   calf   serum   (FCS)   and   antibiotics   were   from   Gibco   Invitrogen   (Carlsbad,  CA).    

2.1.

Animal  experiments.    

The  protocol  for  animal  experiments  was  approved  by  the  University  of  Navarra   Animal   Experimentation   Ethics   Committee   (No.   of   protocol:   060   –   06).   BALB/c   mice  (20  g)  were  obtained  from  Harlan  Interfauna  Ibérica  S.L.  (Barcelona,  Spain).   Animals  received  a  standard  diet  and  water  ad  libitum,  except  for  the  animals  who   received   the   oral   doses,   which   were   fasted   for   24   hours   prior   to   administration.   Female   CB17-­‐SCID   mice   were   purchased   from   Charles   River   Laboratories   (Lyon,   France).   In   xenograft   experiments,   eight-­‐week   SCID   mice   were   subcutaneously   inoculated  into  the  lower  dorsum  with  1  x  107  Z-­‐138  cells  in  100  μL  of  PBS  and  100   μL  of  Matrigel  basement  membrane  matrix  (Becton  Dickinson,  San  Jose,  CA).    

2.2.

Cell  culture  and  isolation  of  human  peripheral  blood  lymphocytes.    

The   Z-­‐138   MCL   cell   line   [19]   was   grown   in   RPMI-­‐1640   culture   medium   supplemented   with   10   %   FCS,   2   mM   L-­‐glutamine,   100   U/mL   penicillin,   and   100   μg/mL  streptomycin  at  37  °C  in  humidified  95  %  air  and  5  %  CO2.   Lymphocytes   were   isolated   from   fresh   human   peripheral   blood   by   dextran   sedimentation,   centrifugation   on   Ficoll-­‐Paque   density   gradients,   and   monocyte   depletion  by  culture  dish  adherence  as  described  previously  [20].        

37  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

2.3.

Edelfosine  uptake  in  cell  culture.    

Drug   uptake   was   measured   as   described   previously   [5]   after   incubating   cells   (106)  with  10  nmol  [3H]edelfosine  for  1  h  in  RPMI-­‐1640/10  %  FCS,  and  subsequent   exhaustive   washing   (six   times)   with   PBS   +   2   %   BSA.   [3H]edelfosine   (specific   activity,   42   Ci/mmol)   was   synthesised   by   tritiation   of   the   9-­‐octadecenyl   derivative   (Amersham  Buchler,  Braunschweig,  Germany).    

2.4.

Apoptosis  assay.  

 Quantitation   of   apoptotic   cells,   following   treatment   with   edelfosine,   was   calculated   by   flow   cytometry   as   the   percentage   of   cells   in   the   sub-­‐G1   region   (hypodiploidy)  in  cell  cycle  analysis,  as  previously  described  [14].    

2.5.

Pharmacokinetic  studies  after  oral  administration.    

Three   treatment   lines   were   followed.   A   BALB/c   mice   group   and   a   SCID   non-­‐ tumour-­‐bearing   mice   group   were   treated   with   a   daily   oral   administration   of   30   mg/kg   of   edelfosine   for   6   days.   At   various   time   points   after   the   first   oral   administration   (0,   1,   2,   5,   8   and   24   h),   blood   was   collected   in   EDTA   surface-­‐coated   tubes   and   then   centrifuged   at   2,000   ×   g   for   10   min   (4   °C)   to   collect   plasma   (100   μL).  After  the  administration  of  the  sixth  dose,  blood  samples  were  collected  at  the   same   time   intervals   (0,   1,   2,   5,   8   and   24   h).   Then,   animals   were   sacrificed   and   spleen,  liver,  lungs,  kidneys,  heart,  brain,  stomach  and  intestine  were  collected  and   weighed.   Tissues   were   homogenised   in   1   mL   of   PBS   pH=7.4   using   a   Mini-­‐bead   Beater   (BioSpect   Products,   Inc.,   Bartelsville,   Oklahoma,   USA)   and   centrifuged   at   10,000   ×   g   for   10   min.   Both   plasma   and   tissue   supernatants   were   collected   and   stored   at   −80   °C   until   high-­‐performance   liquid   chromatography-­‐mass   spectrometry   (HPLC-­‐MS)   analysis   was   performed.     The   last   group,   MCL-­‐   bearing   SCID   mice,   received   daily   oral   administration   of   30   mg/kg   of   edelfosine   for   6   days.   After   the   administration   of   the   sixth   dose,   blood   samples   were   collected   at   the   same  time  intervals  (0,  1,  2,  5,  8  and  24  h),  and  then  the  animals  were  sacrificed   and   liver   and   kidneys   were   collected,   weighed   and   processed   as   previously   described.  Tumours  were  collected  as  the  other  organs.  

38  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

2.6.

Pharmacokinetic  study  after  intravenous  administration.  

 An  intravenous  single  dose  of  200  μg  (10  mg/kg)  was  administered  to  BALB/c   mice  via  the  tail  vein.  At  various  time  points  after  administration  (0,  1,  2,  5,  8  and   24   h),   blood   was   collected   in   EDTA   surface-­‐coated   tubes   and   then   centrifuged   at   2,000   ×   g   for   10   min   (4   °C)   to   separate   the   plasma   (100   μL).   After   24   h   animals   were   sacrificed   by   cervical   dislocation   and   tissueswere   collected,   weighed   and   processed  as  explained  before.      

2.7.

Linearity  study  of  the  dose.    

BALB/c  mice  were  administered  single  intravenous  doses  of  100,  200,  250  and   600  μg  (5,  10,  12.5  and  30  mg/kg)  of  edelfosine  dissolved  in  PBS  via  the  tail  vein.   At  various  time  points  after  administration  (0,  0.5,  1  and  2  h),  blood  was  collected   as  described  above.    

2.8.

Plasma/tissue  extraction  procedure  for  analytical  process.  

10  μg  of  Platelet  Activating  Factor  (0.2  mg/mL)  used  as  internal  standard  were   added   to   100   μl   of   plasma   or   tissue   supernatant.   190   μl   of   a   mixture   of   1   %   formic   acid/methanol   (5:95,   v/v)   were   added   in   order   to   precipitate   the   proteins.   Samples   were   vortexed   for   1   min   and   after   centrifugation   (20,000   ×   g,   10   min),   25   μl  of  the  supernatant  were  analysed  by  HPLC-­‐MS  [18].    

2.9.

HPLC-­‐MS  analysis  for  edelfosine.    

The   method   used   for   edelfosine   quantitation   was   a   slight   modification   of   an   earlier   HPLC-­‐MS   method   [18].   Quantitation   was   achieved   by   comparing   the   observed  peak  area  ratios  of  edelfosine  and  internal  standard  of  the  samples  to  a   regression   curve   determined   from   drug-­‐fortified   plasma   and   tissue   standards.   The   calibration   curve   of   edelfosine   in   plasma   showed   good   linearity   over   the   concentration  range  of  0.1  –  75  μg/mL.  Linear  range  of  edelfosine  concentrations   in  lung,  kidney,  liver,  spleen,  brain,  heart,  stomach  and  intestine  homogenate  was   0.2  –  31.75  μg/mL.     39  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

2.10. Data  analysis.     The   plasma   concentration   data   were   analysed   by   non-­‐compartmental   and   compartmental   analysis   using   WinNonlin   Professional   Edition   Version   2.1   (Pharsight,   Mountain   View,   CA,   USA).   Pharmacokinetic   analysis   was   performed   for   two   different   doses   of   10   and   30   mg/kg   with   plasma   samples   obtained   from   experiments  with  all  mice.  The  area  under  the  plasma  concentration  vs.  time  curve   (AUC)   was   determined   using   the   log-­‐linear   trapezoidal   rule   with   extrapolation   to   24  hours.  Clearance  (CL)  is  the  volume  of  plasma  completely  cleared  of  a  specific   compound  per  unit  time  by  the  organism;  it  was  calculated  by  dividing  the  dose  by   AUC.  The  maximum  plasma  concentration  (Cmax)  was  determined  directly  from  the   plasma  concentration-­‐time  curve.  Oral  bioavailability  (F)  was  determined  by  ratio   of   the   dose-­‐normalized   AUCs   following   oral   and   i.v.   administration.   Volume   of   distribution   at   steady-­‐state   (Vss)   is   the   volume   of   fluid   that   would   be   required   to   contain   the   amount   of   drug   in   the   body   if   it   were   uniformly   distributed   at   a   concentration   equal   to   that   in   the   plasma.   The   half-­‐life   value   (t1/2)   refers   to   the   time   taken   for   plasma   concentration   to   fall   by   50   %.   Disposition   (t1/2α)   and   elimination   (t1/2β)   half-­‐lives   were   determined   using   the   following   formulas:     t1/2α=ln   (2)/α   and   t1/2β=ln   (2)/β,   where   α   and   β   represent   the   disposition   and   elimination   constant   rates,   respectively,   calculated   from   the   intercompartmental   mass  transfer  rates  (k12,  k21)  and  elimination  rates  (k10).    

2.11. Statistical  analysis.     Mean   values   of   the   tissue/plasma   concentration   ratio   of   more   than   2   groups   were  analysed  by  an  analysis  of  variance  (ANOVA)  followed  by  Dunnett's  test  for  a   double   comparison   using   Social   Package   of   Statistical   Sciences   (SPSS).   The   presence   of   differences   in   tissue/plasma   ratios   was   measured   by   the   Mann   Whitney   U   test   for   double   comparisons   using   the   same   program.   A   correlation   analysis   was   performed   for   the   study   of   the   linearity   of   the   dose.   A   value   of   p   <   0.05  was  considered  to  be  statistically  significant  for  all  statistical  tests.        

40  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

3. Results   3.1.

Edelfosine  uptake  and  induction  of  apoptosis  in  MCL  cells.    

Edelfosine   has   been   reported   to   be   selectively   incorporated   into   malignant   cells   leading  to  their  demise,  while  normal  cells  were  spared  [5,  8].  We  found  that  the  Z-­‐ 138   MCL   cell   line   took   up   significant   amounts   of   edelfosine   and   underwent   apoptosis,   whereas   normal   resting   peripheral   blood   lymphocytes   were   not   affected  and  drug  incorporation  was  scarce  (Fig.  1).    

  Figure   1.   Selective   uptake   and   induction   of   apoptosis   in   mantle   lymphoma   cells.   (A)   Edelfosine   uptake  was  determined  after  incubating  human  mantle  lymphoma  Z-­‐138  cells  and  resting  human   peripheral   blood   lymphocytes   (PBL)   with   10   nmol   [3H]edelfosine   for   1   h.   (B)   Z-­‐138   cells   and   resting   human   PBLs   were   incubated   for   24   h   with   10   µM   edelfosine,   and   apoptosis   was   then   quantitated  as  the  percentage  of  cells  in  the  sub-­‐G1  region  in  cell  cycle  analysis  by  flow  cytometry.    

3.2.

Tissue  distribution  of  edelfosine  in  tumour-­‐bearing  SCID  mice.    

Because  Z-­‐138  MCL  cells  incorporated  edelfosine,  we  next  analysed  the  in  vivo   tissue   distribution   of   the   drug   in   tumour-­‐bearing   mice.   Immune   deficient   SCID   mice   were   injected   subcutaneously   with   Z-­‐138   MCL   cells,   and   xenografts   were   allowed  to  establish  to  an  average  size  of  300  mm3  before  drug  oral  treatment.  The   tissue   distribution   expressed   as   tissue/plasma   ratio   of   edelfosine   concentrations   after  multiple  oral  dose  administration  of  an  edelfosine  dose  of  30  mg/kg  (six-­‐day   treatment)  to  MCL-­‐bearing  SCID  mice  is  shown  in  Fig.  2A.     This   oral   30   mg/kg   dose   was   perfectly   well   tolerated   by   the   mice,   and   we   observed   no   side-­‐effects   or   body   weight   loss   (data   not   shown).   Mean   concentration  of  edelfosine  in  plasma  24  h  after  the  sixth  dose  was  10.69  μg/mL.   41  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

Kidney  and  liver  were  selected  for  drug  accumulation,  as  they  are  the  major  drug   clearance   tissues.   These   organs   showed   a   two-­‐fold   higher   drug   concentration   than   the  corresponding  one  in  plasma  (Fig.  2A).  Interestingly,  the  tumour  showed  a  high   accumulation   of   edelfosine   24   h   after   the   dose   on   day   6   (Fig.   2A).   The   mean   concentration  of  edelfosine  in  the  tumour  at  this  stage  was  138.64  μg/g,  which  is   about   thirteen   times   higher   than   the   plasma   drug   concentration,   and   five   to   six   times  higher  than  the  drug  concentration  found  in  kidney  and  liver.  Moreover,  the   tumour/plasma   concentration   ratio   of   edelfosine   was   significantly   higher   (p   <   0.01)  than  the  corresponding  ratios  observed  in  both  kidney  and  liver.  Thus,  an  in   vivo   and   in   vitro   comparison   indicates   that   edelfosine   is   selectively   taken   up   by   tumour  cells.  

  Figure   2.   A)   Tissue/plasma   concentration   ratios   of   edelfosine   after   (   )   multiple   dose   oral   administration   of   30   mg/kg   of   edelfosine   for   six   days   to   non   tumor-­‐bearing   SCID   mice   and   (       )   multiple  dose  oral  administration  of  30  mg/kg  of  edelfosine  for  six  days  to  mantle  cell  lymphoma   bearing  SCID  mice  (n=6,  Mean  ±  S.D.);  and  B)  tissue/plasma  concentration  ratios  of  edelfosine  after   (    )  single  intravenous  dose  of  10  mg/kg  of  edelfosine  to  healthy  BALB/c  mice;  (      )  multiple  dose   oral  administration  of  30  mg/kg  of  edelfosine  for  six  days  to  healthy  BALB/c  mice;    (      )  multiple   dose   oral   administration   of   30   mg/kg   of   edelfosine   for   six   days   to   non   tumor-­‐bearing   SCID   mice   (n=6,  Mean  ±  S.D.)  

  42  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

3.3.

Tissue  distribution  of  edelfosine  in  non-­‐tumour-­‐bearing  SCID  mice.    

The  tissue  distribution  of  edelfosine  was  also  analysed  in  control  non-­‐tumour-­‐ carrying  SCID  mice  following  the  same  protocol  as  with  tumour-­‐bearing  SCID  mice.   The   mean   plasma   concentration   of   edelfosine   at   the   time   of   24   h   after   the   sixth   dose   was   12.12   μg/mL.   Fig.   2B   shows   a   higher   distribution   of   edelfosine   to   lung,   spleen,  intestine,  liver  and  kidney.  In  contrast,  low  drug  concentrations  were  found   in  heart,  brain  and  stomach  (Fig.  2B).  Compared  to  tumour-­‐bearing  SCID  mice,  no   statistical  differences  in  liver/plasma  concentration  ratios  (p  >  0.05)  were  found  in   non-­‐tumour-­‐bearing   SCID   mice,   whereas   the   kidney   was   found   to   have   a   statistically  highly  significant  increase  (p  <  0.01)  (Fig.  2A).     3.4.

Tissue  distribution  in  healthy  BALB/c  mice  after  oral  administration.    

A   multiple   oral   dose   administration   of   30   mg/kg   of   edelfosine   (six-­‐day   treatment)   was   performed   to   healthy   BALB/c   mice.   24   h   after   the   last   dose   (day   six),  the  mean  plasma  concentration  was  13.22  μg/mL.  As  is  shown  in  Fig.  2B,  the   highest   tissue/plasma   concentration   ratios   were   found   for   kidney   and   intestine.   Lower  ratios  were  observed  for  stomach,  spleen,  lung  and  liver.  A  low  amount  of   edelfosine  was  detected  in  the  heart  and  brain.     Fig.  2B  also  shows  that  no  statistically  significant  differences  were  appreciated   among   the   tissue/plasma   concentration   ratios   of   lung,   heart,   brain,   intestine   and   liver  of  BALB/c  and  non-­‐tumour-­‐bearing  SCID  mice.  The  stomach  of  BALB/c  mice   after   the   multiple   oral   administration   of   30   mg/kg   presented   a   higher   ratio   than   SCID   mice   after   the   administration   of   same   dose   (p   <   0.05)   and   the   spleen/plasma   concentration   ratio   showed   a   significant   decrease   (p   <   0.05)   with   regard   to   SCID   non-­‐tumour-­‐bearing  mice.       3.5.

Tissue   distribution   in   healthy   BALB/c   mice   after   intravenous  

administration.     A   single   intravenous   administration   of   200   μg   of   edelfosine   (10   mg/kg)   was   administered   to   healthy   BALB/c   mice.   After   24   h,   edelfosine   concentration   in   43  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

plasma  was  around  2.5  μg/mL.  The  tissue  distribution  of  edelfosine  expressed  as   tissue/plasma   ratio   can   be   observed   in   Fig.   2B.   No   drug   was   detectable   in   heart   and   very   little   was   found   in   brain.   Kidney   and   intestine   presented   the   highest   tissue/plasma  concentration  ratios,  followed  by  lung,  liver,  stomach  and  spleen.     3.6.

Pharmacokinetic  characterization  after  intravenous  administration.  

 Fig.  3  depicts  the  concentration  of  edelfosine  in  mouse  plasma  plotted  against   time   after   a   single   dose   intravenous   administration   of   200   μg   of   edelfosine   (10   mg/kg)  to  BALB/c  mice.    

  Figure   3.   Time-­‐concentration   curve   data   of   edelfosine   24   h   after   single   dose   intravenous   (♦)   administration  of  10  mg/kg  and  oral  single  dose  (▲)  administration  of  30  mg/kg  to  BALB/c  mice   (n=6,  Mean  ±  S.D.)    

Pharmacokinetic   analysis   of   edelfosine   in   blood   plasma   showed   a   Cmax   of   50.7±28.1   μg/mL   and   a   Cmin   of   2.5±1.3   μg/mL,   24   hours   after   intravenous   administration.  The  obtained  pharmacokinetic  parameters  are  listed  in  Table  I.      

44  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

Table  I.  Comparison  of  pharmacokinetic  parameters  of  edelfosine  after  administration  of  different   doses   to   BALB/c   and   SCID   mice   by   different   routes   (n=6,   Mean   ±   S.D.).   No   statistical   differences   were  found  among  the  parameters  (p  >  0.05)  

  Plasma   concentration-­‐time   data   of   edelfosine   was   well   described   by   a   bi-­‐ exponential   function   (model   selection   criterion,   -­‐24.12)   following   intravenous   administration.   The   half-­‐lives   of   distribution   (t1/2α)   and   elimination   (t1/2β)   phases   were   0.286±0.076   and   22.288±14.016   h,   respectively.   The   systemic   clearance   (CL)   and   steady-­‐state   volume   of   distribution   (Vss)   were   0.056±0.030   L/h/kg   and   1.285±0.556   L/kg,   respectively.   There   was   little   variability   in   most   of   the   values   of   the  parameters,  indicating  a  well-­‐controlled  and  reproducible  study,  except  for  the   elimination  phase  half-­‐life  value.       3.7.

Linearity  of  dose.    

Both  AUC-­‐dose  and  Cmax-­‐dose  profiles  of  edelfosine  showed  a  linear  correlation   (r2=0.999),   as   can   be   seen   in   Fig.   4,   suggesting   linear   pharmacokinetics   with   similar  elimination  and  distribution  half-­‐lives  after  intravenous  administration  of   doses  between  5  and  30  mg/kg.    

45  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

  Figure  4.  Correlation  between  doses  of  edelfosine  of  100,  200,  250  and  600  µg  (5,  10,  12.5  and  30   mg/kg)  and  the  calculated  AUC  (A)  and  Cmax  (B)  (n=6,  Mean  ±  S.D.)    

3.8.

Pharmacokinetic  characterization  after  oral  administration.    

A  very  low  concentration  of  edelfosine  could  be  found  in  plasma  along  the  time   interval   of   0,   1,   2,   5,   8   and   24   h   after   oral   administration   of   30   mg/kg   of   edelfosine   in  BALB/c  mice,  as  can  be  observed  in  Fig.  3.  These  concentrations  were  close  to   the   detectable   limits   of   the   technique   and   not   sufficient   for   the   calculation   of   pharmacokinetic   parameters.   Oral   bioavailability   for   edelfosine   was   calculated   from   the   ratios   of   the   average   values   for   AUC0→24h   for   the   oral   and   intravenous   doses.  Oral  bioavailability  (F)  for  edelfosine  was  found  to  be  less  than  10  %.   Pharmacokinetic   parameters   obtained   after   compartmental   analysis   of   experimental   data   are   shown   in   Table   I.   Oral   bioavailability   increased   to   64   %,   when   the   steady-­‐state   was   reached   after   the   sixth   day.   At   this   point,   a   Cmax   of   14.46±2.97  μg/mL  at  a  tmax  of  3  h  and  a  Cmin  of  7.65±2.45  μg/mL  were  measured  24   hours   after   the   last   administration.   Due   to   the   fast   elimination   during   the   disposition   phase,   only   the   β   elimination   phase   could   be   characterized.   AUC   presented   a   value   of   258.54±58.16   μg·h/L.   The   half-­‐life   of   elimination   was   46  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

30.404±26.761   h,   with   a   volume   of   distribution   at   steady-­‐state   of   1.563±0.163   L/kg   and   a   systemic   clearance   of   0.057±0.039   L/h/kg,   similar   values   to   those   obtained   after   intravenous   administration   of   edelfosine,   with   no   statistically   significant  differences.   In   order   to   determine   possible   inter-­‐strain   pharmacokinetic   differences,   these   results   were   compared   to   the   results   obtained   after   edelfosine   was   administered   to   non   tumour-­‐bearing   SCID   mice.   After   reaching   the   steady-­‐state,   pharmacokinetic   parameters   were   estimated   (Table   I).   Whatever   the   parameters   studied   (half   life   of   elimination,   clearance   value,   steady-­‐state   volume   of   distribution   and   AUC)   no   statistically   significant   differences   were   observed   between  SCID  and  BALB/c  mice.    

4. Discussion   Several   attempts   have   been   made   over   the   years   accurately   to   detect   the   accumulation  of  edelfosine  in  tumours,  tissues  and  plasma  as  well  as  to  estimate  its   pharmacokinetic  parameters.  This  pharmacokinetic  analysis  is  mandatory  in  order   to  accomplish  all  the  preclinical  assays  prior  to  a  rational  clinical  use  of  the  drug.   Because   edelfosine   shows   a   variety   of   medical   applications,   a   pharmacokinetic   study  in  different  murine  species,  each  one  appropriate  for  distinct  in  vivo  assays,   is   required.   We   carried   out   a   complete   pharmacokinetic   analysis   in   SCID   mice,   widely   used   for   anti-­‐tumour   activity   testing,   and   BALB/c,   useful   for   additional   putative   edelfosine   applications   [21].   In   the   present   study,   we   set   up   a   MCL-­‐ bearing  animal  model  in  immune  deficient  SCID  mice  to  analyse  the  distribution  of   edelfosine   in   tissues   and   tumour.   We   found   that   the   Z-­‐138   MCL   cell   line   took   up   significant   amounts   of   edelfosine,   subsequently   undergoing   apoptosis,   whereas   normal  cells  hardly  incorporated  the  drug  and  were  spared.  The  in  vitro  and  in  vivo   data   reported   here   indicate   a   remarkably   selective   accumulation   of   edelfosine   in   tumour  cells.   In   the   MCL-­‐bearing   animal   model,   kidney   and   liver   were   the   only   organs   extracted   as   they   are   considered   to   be   the   main   drug   distribution   and   clearance   tissues.   Comparing   the   tissue   distribution   of   edelfosine   between   these   tumour-­‐ 47  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

bearing  SCID  mice  and  the  non-­‐tumour-­‐bearing  SCID  mice  (Fig.  2A),  no  statistically   significant   differences   were   seen   in   liver   (p   >   0.05),   while   a   major   decrease   (p   <   0.01)   was   found   in   the   kidneys   of   the   tumour-­‐bearing   mice.   This   lower   renal   uptake   in   the   tumor   model   might   be   a   result   of   an   increased   drug   uptake   by   the   tumor  with  less  drug  available  for  renal  excretion,  in  addition  to  the  renal  failure   usually   expected   at   advanced   stages   of   the   disease   [22].   These   results   clearly   indicate  that  MCL  does  not  seem  to  affect  the  tissue  distribution  of  the  drug.   Fig.   2B   displays   that   the   distribution   of   edelfosine   in   mice   is   mainly   predominant   in   spleen,   intestine   and   kidney.   The   high   presence   of   edelfosine   in   spleen   can   be   explained   by   the   incorporation   of   the   drug   into   the   membrane   of   erythrocytes  inducing  a  haemolytic  effect  [17].  These  red  blood  cells  end  up  being   cleared   from   the   blood   in   the   spleen.   We   found   a   higher   concentration   of   edelfosine  in  the  spleen  of  SCID  mice  as  compared  to  BALB/c  mice,  and  this  could   be  due  to  putative  differences  in  the  spleen  of  immune  deficient  mice.   No   statistically   significant   differences   were   observed   between   tissue/plasma   concentration  ratios  of  edelfosine  in  lung,  heart,  brain,  intestine  liver  and  kidney  of   immune   suppressed   mice   (SCID   mice)   and   BALB/c   mice   (Fig.   2B).   Moreover,   no   significant  differences  were  observed  in  mean  plasma  concentrations  of  edelfosine   24   h   after   the   sixth   dose   of   the   multiple   oral   administration   of   30   mg/kg   to   tumour-­‐bearing   SCID   mice,   non-­‐tumour-­‐bearing   SCID   mice   and   healthy   BALB/c   mice   (10.69   μg/mL,   12.12   μg/mL   and   13.22   μg/mL,   respectively).   As   a   result,   a   similar  pharmacokinetic  profile  would  be  expected  in  both  strains.  Therefore,  we   performed   the   characterisation   of   edelfosine   pharmacokinetic   profile   in   healthy   BALB/c  mice.   Knowledge   of   the   pharmacokinetic   parameters   of   a   drug   is   a   must   in   order   to   ensure  that  exposure  is  sufficient  to  evaluate  its  pharmacodynamic  properties,  to   predict   the   pharmacokinetics   in   other   species   and   to   develop   an   appropriate   dosage  form.     Following   intravenous   administration   of   a   dose   of   10   mg/kg   of   edelfosine   to   BALB/c   mice,   mean   blood   concentrations   declined   biphasically,   with   an   initial   half   life   of   around   0.3   h   (Table   I).   After   2   h,   plasma   concentrations   appeared   to   decline   48  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

more   slowly.   Given   that   blood   concentrations   had   declined   about   six-­‐fold   during   the   initial   phase,   the   contribution   of   the   secondary   phase   to   the   total   drug   exposure   is   of   little   relevance.   According   to   these   half-­‐life   values,   edelfosine   presented   a   rapid   distribution   to   central   compartment   tissues   whereas   a   slower   distribution   to   deep   compartments   was   appreciated.   The   rapid   distribution   is   mostly   observed   to   organs   that   are   more   highly   irrigated,   like   kidney,   liver,   intestine  and  lung.     Steady-­‐state  volume  of  distribution  was  1.26  L/kg,  much  greater  than  that  of  the   vascular  volume  in  mice  and  approximately  twice  its  total  body  water  [23],  when   normalized  to  body  weight,  suggesting  that  edelfosine  is  highly  distributed  extra-­‐ vascularly.   In   fact,   k12   presented   a   value   five   to   six   times   higher   than   the   corresponding  k21  value.     Edelfosine  showed  very  low  clearance  values  in  mice  compared  to  its  respective   liver  and  kidney  blood  flow.  It  is  interesting  to  note  that  this  value  of  0.056  L/h/kg   is   translated   to   barely   1   %   of   the   liver   blood   flow  [23],   suggesting   a   much   lower   intrinsic  hepatic  clearance  value.     We   verified   that   as   the   drug   dose   increases   the   area   under   the   curve   (AUC)   increases   equivalently,   meaning   that   edelfosine   exhibits   linear   pharmacokinetics.   In   this   study   we   demonstrate   that   edelfosine   shows   a   linear   correlation   not   only   between   the   administered   dose   and   the   resulting   AUC,   but   also   between   the   administered  dose  and  the  corresponding  Cmax.  This  fact  suggests  that  there  is  no   saturation   of   the   elimination   process   of   edelfosine   at   the   concentration   range   between  5  and  30  mg/kg.     Even   though   the   mechanism   for   its   clearance   is   still   unknown,   renal   elimination   of   the   drug   might   well   happen,   as   edelfosine   is   present   in   kidney.   Previous   studies   have   established   that   amphiphilic   cationic   drugs   are   efficiently   removed   by   the   liver  from  the  blood  [24].  Additionally,  amphiphilic  cationic  drugs  were  detected  in   other   secretion   organs   of   cationic   amphiphilic   drugs   like   intestinal   mucosa   [25]   and  kidney.  These  data  are  also  in  good  agreement  with  our  results  and  the  ones   obtained  by  Marschner  et  al.  [26].    

49  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

The  first  determinations   of   oral   absorption  of  15  μmol  of  edelfosine  (38  mg/kg)   in  1992  suggested  that  about  96  %  of  edelfosine  was  absorbed  by  rats  within  24   hours   [17].   Conversely,   our   study   shows   very   low   plasma   concentrations   of   edelfosine   24   hours   after   the   oral   administration   of   a   single   dose   of   30   mg/kg,   revealing   very   low   absorption   of   edelfosine   by   the   gastrointestinal   tract   in   mice.   However,   oral   bioavailability   showed   a   significant   increase   (64   %)   after   daily   administration  of  30  mg/kg  of  edelfosine  for  6  days.   As   can   be   seen   from   our   studies,   edelfosine   showed   a   moderate   volume   of   distribution   and   a   rapid   and   varied   distribution   through   the   organism   when   it   is   administered   by   an   intravenous   route.   This   information,   along   with   the   amphiphilic   properties,   may   lead   us   to   consider   the   possibility   of   passive   diffusion   through   the   intestinal   epithelium   as   mechanism   of   absorption   for   edelfosine.   However,   this   suggestion   does   not   correlate   with   the   low   absorption   rate   of   the   drug  found  using  the  oral  route.  Moreover,  it  has  been  shown  that  the  absorption   of   certain   cationic   drugs,   such   as   quaternary   ammonium   compounds,   shows   a   reduced   absorption   rate   due   to   the   presence   of   efflux   transport   systems   on   the   apical  membrane  (e.g.  P-­‐gp).  Indeed,  the  alkylphosphocholine  miltefosine  has  been   reported  to  interact  with  P-­‐gp  [27],  and  it  has  also  been  reported  that  edelfosine  is   a   substrate   for   P-­‐glycoprotein   [28].   This   issue   would   only   explain   the   lack   of   absorption  at  a  single  oral  dose,  and  the  high  oral  bioavailability  achieved  after  a   multiple  oral  dose  administration.     In   summary,   edelfosine   showed   a   remarkable   apoptotic   effect   in   Z-­‐138   MCL   cells   in   vitro   and   a   high   and   rather   selective   uptake   in   MCL   tumour   cells   in   vitro   and   in   vivo.   This   fact,   along   with   knowledge   of   the   biodistribution   and   pharmacokinetic  behaviour  of  edelfosine,  permits  the  establishment  of  treatment   regimens   as   well   as   the   development   of   drug   delivery   systems   in   order   to   treat   MCL   and   other   tumours.   In   addition,   since   no   statistically   significant   differences   were  noticed  among  the  pharmacokinetic  parameters  of  all  three  treatment  lines,   it   can   be   concluded   that   there   are   no   differences   between   BALB/c   and   SCID   pharmacokinetic   profiles.   Thus,   further   studies   with   edelfosine   delivery   systems   will  be  able  to  be  performed  either  in  one  animal  model  or  the  other.  

50  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

These  data  indicate  that  multiple  oral  administration  of  edelfosine  is  required  to   reach   a   clinically   important   plasma   concentration.   Our   results   also   show   that   edelfosine   is   widely   scattered   across   different   organs,   but   that   it   is   preferentially   internalised  by  the  tumour  both  in  vitro  and  in  vivo.    

Financial  support     Caja   Navarra   Foundation,   the   Spanish   Ministry   of   Science   and   Innovation   (SAF2007-­‐61261,   SAF2005-­‐04293,   PCT-­‐090100-­‐2007-­‐27)   the   “Fondo   de   Investigación   Sanitaria”   and   European   Commission   (FIS-­‐FEDER   06/0813)   and   RD06/0020/1037   from   Red   Temática   de   Investigación   Cooperativa   en   Cáncer   (RTICC),   Instituto   de   Salud   Carlos   III   (ISCIII),   Ministerio   de   Sanidad.   Consuelo   Gajate   is   supported   by   the   Ramón   y   Cajal   Program   from   the   Spanish   Ministry   of   Science   and   Innovation.   Ander   Estella-­‐Hermoso   de   Mendoza   is   supported   by   the   research   grant   from   the   Department   of   Education   of   the   Basque   Government   (BFI06.37).  

51  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

References     1.  

Gajate,   C.   and   F.   Mollinedo,   Biological   Activities,   Mechanisms   of   Action   and   Biomedical   Prospect   of   the   Antitumor   Ether   Phospholipid   ET-­‐18-­‐OCH3   (Edelfosine),   A   Proapoptotic   Agent   in   Tumor   Cells.   Curr   Drug   Metab,   2002.   3(5):  p.  491-­‐525.  

2.  

Mollinedo,   F.,   et   al.,   ET-­‐18-­‐OCH3   (edelfosine):   a   selective   antitumour   lipid   targeting   apoptosis   through   intracellular   activation   of   Fas/CD95   death   receptor.  Curr  Med  Chem,  2004.  11(24):  p.  3163-­‐84.  

3.  

Houlihan,   W.J.,   et   al.,   Phospholipid   antitumor   agents.   Med.   Res.   Rev.,   1995.   15(3):  p.  157-­‐223.  

4.  

Munder,  P.G.  and  O.  Westphal,  Antitumoral  and  other  biomedical  activities  of   synthetic  ether  lysophospholipids.  Chem.  Immunol.,  1990.  49:  p.  206-­‐35.  

5.  

Mollinedo,   F.,   et   al.,   Selective   induction   of   apoptosis   in   cancer   cells   by   the   ether   lipid   ET-­‐   18-­‐OCH3   (Edelfosine):   molecular   structure   requirements,   cellular   uptake,   and   protection   by   Bcl-­‐2   and   Bcl-­‐X(L).   Cancer   Res,   1997.   57(7):  p.  1320-­‐8.  

6.  

Ruiter,   G.A.,   et   al.,   Alkyl-­‐lysophospholipids   as   anticancer   agents   and   enhancers  of  radiation-­‐  induced  apoptosis.  Int  J  Radiat  Oncol  Biol  Phys,  2001.   49(2):  p.  415-­‐9.  

7.  

Gajate,   C.   and   F.   Mollinedo,   Edelfosine   and   perifosine   induce   selective   apoptosis   in   multiple   myeloma   by   recruitment   of   death   receptors   and   downstream  signaling  molecules  into  lipid  rafts.  Blood,  2007.  109(2):  p.  711-­‐ 9.  

8.  

Gajate,   C.,   et   al.,   Intracellular   triggering   of   Fas,   independently   of   FasL,   as   a   new   mechanism   of   antitumor   ether   lipid-­‐induced   apoptosis.   Int   J   Cancer,   2000.  85(5):  p.  674-­‐82.  

52  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

9.  

Gajate,   C.   and   F.   Mollinedo,   The   antitumor   ether   lipid   ET-­‐18-­‐OCH3   induces   apoptosis   through   translocation   and   capping   of   Fas/CD95   into   membrane   rafts  in  human  leukemic  cells.  Blood,  2001.  98(13):  p.  3860-­‐3.  

10.  

Gajate,   C.,   et   al.,   Intracellular   Triggering   of   Fas   Aggregation   and   Recruitment   of   Apoptotic   Molecules   into   Fas-­‐enriched   Rafts   in   Selective   Tumor   Cell   Apoptosis.  J  Exp  Med,  2004.  200(3):  p.  353-­‐65.  

11.  

Gajate,  C.,  et  al.,  Involvement  of  c-­‐Jun  NH2-­‐terminal  kinase  activation  and  c-­‐ Jun  in  the  induction  of  apoptosis  by  the  ether  phospholipid  1-­‐O-­‐octadecyl-­‐2-­‐O-­‐   methyl-­‐rac-­‐glycero-­‐3-­‐phosphocholine.   Mol   Pharmacol,   1998.   53(4):   p.   602-­‐ 12.  

12.  

Ruiter,   G.A.,   et   al.,   Alkyl-­‐lysophospholipids   activate   the   SAPK/JNK   pathway   and   enhance   radiation-­‐induced   apoptosis.   Cancer   Res,   1999.   59(10):   p.   2457-­‐63.  

13.  

Nieto-­‐Miguel,   T.,   et   al.,   Endoplasmic   Reticulum   Stress   in   the   Proapoptotic   Action   of   Edelfosine   in   Solid   Tumor   Cells.   Cancer   Res,   2007.   67(21):   p.   10368-­‐10378.  

14.  

Gajate,  C.,  et  al.,  Involvement  of  mitochondria  and  caspase-­‐3  in  ET-­‐18-­‐OCH3-­‐ induced   apoptosis   of   human   leukemic   cells.   Int   J   Cancer,   2000.   86(2):   p.   208-­‐ 18.  

15.  

Vogler,   W.R.,   et   al.,   A   phase   II   trial   of   autologous   bone   marrow   transplantation   (ABMT)   in   acute   leukemia   with   edelfosine   purged   bone   marrow.  Adv  Exp  Med  Biol,  1996.  416:  p.  389-­‐96.  

16.  

Arnold,   B.,   R.   Reuther,   and   H.U.   Weltzien,   Distribution   and   metabolism   of   synthetic  alkyl  analogs  of  lysophosphatidylcholine  in  mice.  Biochim.  Biophys.   Acta.,  1978.  530(1):  p.  47-­‐55.  

17.  

Kotting,   J.,   et   al.,   Hexadecylphosphocholine   and   octadecyl-­‐methyl-­‐glycero-­‐3-­‐ phosphocholine:   a   comparison   of   hemolytic   activity,   serum   binding   and   tissue   distribution.  Prog.  Exp.  Tumor  Res.,  1992.  34:  p.  131-­‐42.  

18.  

Blanco-­‐Prieto,   M.J.,   M.A.   Campanero,   and   F.   Mollinedo,   Quantitative   determination   of   the   antitumor   alkyl   ether   phospholipid   edelfosine   by   53  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

reversed-­‐phase   liquid   chromatography-­‐electrospray   mass   spectrometry:   application   to   cell   uptake   studies   and   characterization   of   drug   delivery   systems.  J.  Chromatogr.  B.  Analyt.  Technol.  Biomed.  Life  Sci.,  2004.  810(1):   p.  85-­‐92.   19.  

Medeiros,   L.J.,   Z.   Estrov,   and   G.Z.   Rassidakis,   Z-­‐138   cell   line   was   derived   from   a  patient  with  blastoid  variant  mantle  cell  lymphoma.  Leuk  Res,  2006.  30(4):   p.  497-­‐501.  

20.  

Cabaner,   C.,   et   al.,   Induction   of   apoptosis   in   human   mitogen-­‐activated   peripheral   blood   T-­‐   lymphocytes   by   the   ether   phospholipid   ET-­‐18-­‐OCH3:   involvement   of   the   Fas   receptor/ligand   system.   Br   J   Pharmacol,   1999.   127(4):  p.  813-­‐25.  

21.  

Mollinedo,   F.,   Antitumor   ether   lipids:   proapoptotic   agents   with   multiple   therapeutic  indications.  Expert  Opin  Ther  Patents,  2007.  17(4):  p.  385-­‐405.  

22.  

Davies,   J.,   et   al.,   Acute   renal   failure   due   to   mantle   cell   lymphoma-­‐-­‐a   case   report  and  discussion  of  the  literature.  Clin.  Nephrol.,  2007.  67(6):  p.  394-­‐6.  

23.  

Davies,   B.   and   T.   Morris,   Physiological   parameters   in   laboratory   animals   and   humans.  Pharm.  Res.,  1993.  10(7):  p.  1093-­‐5.  

24.  

Smit,   J.W.,   et   al.,   Hepatobiliary   and   intestinal   clearance   of   amphiphilic   cationic   drugs   in   mice   in   which   both   mdr1a   and   mdr1b   genes   have   been   disrupted.  Br.  J.  Pharmacol.,  1998.  124(2):  p.  416-­‐24.  

25.  

Kim,   M.K.   and   C.K.   Shim,   The   transport   of   organic   cations   in   the   small   intestine:   current   knowledge   and   emerging   concepts.   Arch.   Pharm.   Res.,   2006.  29(7):  p.  605-­‐16.  

26.  

Marschner,  N.,  et  al.,  Distribution  of  hexadecylphosphocholine  and  octadecyl-­‐ methyl-­‐glycero-­‐3-­‐phosphocholine  in  rat  tissues  during  steady-­‐state  treatment.   Cancer  Chemother.  Pharmacol.,  1992.  31(1):  p.  18-­‐22.  

27.  

Rybczynska,   M.,   et   al.,   MDR1   causes   resistance   to   the   antitumour   drug   miltefosine.  Br.  J.  Cancer.,  2001.  84(10):  p.  1405-­‐11.  

54  

Chapter  1.  Antitumor  alkyl  ether  lipid  edelfosine:  tissue  distribution  and  pharmacokinetic   behavior  in  healthy  and  tumor-­‐bearing  immunosuppressed  mice    

28.  

Ruetz,   S.,   et   al.,   Functional   interactions   between   synthetic   alkyl   phospholipids   and   the   ABC   transporters   P-­‐glycoprotein,   Ste-­‐6,   MRP,   and   Pgh   1.   Biochemistry,  1997.  36(26):  p.  8180-­‐8.  

55  

 

 

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

   

Chapter  2    

Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine   encapsulation:  development  and  in  vitro  characterization    

Ander  Estella-­‐Hermoso  de  Mendoza1,  Marta  Rayo1,  Faustino  Mollinedo2  and  María  J.   Blanco-­‐Prieto1*     1Department  of  Pharmacy  and  Pharmaceutical  Technology,  School  of  Pharmacy,  

University  of  Navarra,  C/Irunlarrea  1,  E-­‐31080  Pamplona,  Spain   2Centro  de  Investigación  del  Cáncer,  Instituto  de  Biología  Molecular  y  Celular  del  

Cáncer,  CSIC-­‐Universidad  de  Salamanca,  Campus  Miguel  de  Unamuno,  E-­‐37007   Salamanca,  Spain    

Key   words:   Lipid   nanoparticles;   Edelfosine;   Drug   delivery;   Atomic   force   microscopy;  Differential  scanning  calorimetry;  X–ray  diffractometry.   Corresponding   author:   Dr.   María   J.   Blanco-­‐Prieto,   Department   of   Pharmacy   and   Pharmaceutical   Technology,   School   of   Pharmacy,   University   of   Navarra,   C/Irunlarrea   1,   E-­‐31080   Pamplona,   Spain,   Office   phone:   +   34   948   425   600   ext.   6519,  Fax:  +  34  948  425  649,  e-­‐mail:  [email protected]     European  Journal  of  Pharmaceutics  and  Biopharmaceutics,  2008.  68(2):  p.  207-­‐13

57  

 

 

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

 

Abstract     The  

ether  

lipid  

1-­‐O-­‐octadecyl-­‐2-­‐O-­‐methyl-­‐rac-­‐glycero-­‐3-­‐phosphocholine,  

edelfosine   (ET-­‐18-­‐OCH3)   is   the   prototype   molecule   of   a   promising   class   of   antitumour   drugs   named   alkyl–lysophospholipid   analogues   (ALPs)   or   antitumor   ether   lipids.   This   drug   presents   a   very   important   drawback   as   can   be   the   dose   depending   haemolysis   when   administered   intravenously.   Lipid   nanoparticles   have   been   lately   proposed   for   different   drug   encapsulation   as   an   alternative   to   other   controlled  release  delivery  systems,  such  as  liposomes  or  polymeric  nanoparticles.   The   aim   of   this   study   was   to   develop   a   lipid   nanoparticulate   system   that   would   decrease  systemic  toxicity  as  well  as  improve  the  therapeutic  potential  of  the  drug.   Lipids   employed   were   Compritol®   888   ATO   and   stearic   acid.   The   nanoparticles   were   characterized   by   photon   correlation   spectroscopy   for   size   and   size   distribution,   and   atomic   force   microscopy   (AFM)   was   used   for   the   determination   of   morphological   properties.   By   both   differential   scanning   calorimetry   (DSC)   and   X-­‐ray   diffractometry,   crystalline   behaviour   of   lipids   and   drug   was   assessed.   The   drug   encapsulation   efficiency   and   the   drug   release   kinetics   under   in   vitro   conditions   were   measured   by   HPLC–MS.   It   was   concluded   that   Compritol®   presents   advantages   as   a   matrix   material   for   the   manufacture   of   the   nanoparticles   and  for  the  controlled  release  of  edelfosine.  

59  

 

   

 

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

1. Introduction   Edelfosine  

(ET–18–OCH3,  

1-­‐O-­‐octadecyl-­‐2-­‐O-­‐methyl-­‐rac-­‐glycero-­‐3-­‐

phosphocholine)   is   the   prototype   of   a   promising   class   of   antitumor   agents,   collectively   known   as   alkyl-­‐lysophospholipid   analogues   (ALP)   or   antitumor   ether   lipids,  that  do  not  target  the  DNA,  but  affect  the  cell  membrane  and  the  apoptotic   machinery  of  the  cancer  cell  [1].  Phase  I  studies  have  shown  a  good  tolerability  of   the   drug   but   with   haematological   and   systemic   side   effects   [1,   2].   However,   although  edelfosine  has  been  shown  to  exert  potent  antineoplastic  effects  in  vitro   [3,  4],  the  antitumor  activity  in  phase  II  clinical  studies  has  been  only  moderate  [5].   Moreover,  ALPs  show  manifold  biological  effects  in  addition  to  their  antineoplastic   actions,  including  an  antiparasitic  effect  on  Leishmania  [6]  as  well  as  an  inhibition   of   the   cell   membrane   phospholipid   turnover   [7]   and   a   potent   inhibition   of   neovascularization  [8],  protein  kinase  C  and  Na+/K+-­‐ATPase  [9].   Edelfosine  has  also  been  given  intravenously,  but  this  provoked  haemolysis  as  a   major   side   effect   [10].   This   led   to   the   only   formulation   developed   so   far,   the   TLC   ELL-­‐12,   in   which   edelfosine   was   included   into   liposomes   [11]   to   avoid   the   haemolytic  toxicity  of  the  drug.  However,  the  main  inconvenience  of  liposomes  is   their  rapid  clearance  from  plasma  in  comparison  with  other  delivery  systems.   Lipid   nanoparticles   have   been   proposed   as   an   alternative   for   the   existing   traditional   particulate   systems,   such   as   previously   mentioned   liposomes   or   polymeric  nanoparticles.  These  particulate  systems  made  from  solid  lipids  started   being   developed   in   the   early   nineties.   They   provide   physical   stability,   controlled   release   and   a   wide   variety   of   application   routes   (parenteral,   oral,   dermal,   ocular,   pulmonary  and  rectal)  [12-­‐16].     Lipid   nanoparticles   are   basically   composed   of   a   high   melting   point   lipid   that   acts   as   a   solid   core,   covered   by   surfactants.   Lipids   used   to   form   these   matrices   are   biodegradable   raw   materials   that   are   physiologically   tolerated   [17]:   triglycerides   (i.e.   tristearin),   partial   glycerides   (i.e.   Compritol),   fatty   acids   (i.e.   stearic   acid),   steroids  (i.e.  cholesterol)  or  waxes  (i.e.  cetyl  palmitate)  [18].  

61  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

The   formulation   methods   are   also   diverse.   Emulsion   formation   and   solvent   evaporation   method   has   been   widely   used   for   particle   formation,   although   other   methods   like   high   pressure   homogenization   [18],   solvent   diffusion   methods   in   aqueous  solutions  [19]  or  hot  emulsion  methods  [18,  20]  have  also  been  employed.   Drawbacks   associated   to   this   kind   of   formulations,   like   limited   drug   loading   capacity,   adjustment   of   drug   release   profile   and   potential   drug   expulsion   during   storage   have   been   reported   [21].   Besides,   drug   loading   capacity   is   limited   by   the   solubility   of   the   drug   in   the   lipid   melt,   the   structure   of   the   lipid   matrix   and   the   polymorphic  state  of  the  lipid  matrix.   Comparing   with   the   previously   mentioned   liposomes,   the   main   improvement   of   the  lipid  nanoparticles  is  their  physical  and  chemical  long-­‐term  stability  up  to  12  –   24   months   [13],   even   though   an   increase   in   particle   size   has   been   reported   in   a   lesser   time   [22].   As   a   feasible   solution   for   this   setback,   the   freeze-­‐drying   process   has   shown   to   increase   physicochemical   stability   of   lipid   particles   over   large   periods  of  time  [23].   Taking  into  account  this  information,  the  aim  of  this  study  was  to  develop  a  new   formulation   that   would   provide   a   controlled   release   for   the   antitumor   lipid   edelfosine,  in  order  to  improve  its  therapeutic  activity.    

2. Materials  and  Methods   2.1.

Materials    

Edelfosine   was   from   INKEYSA   (Barcelona,   Spain).   Compritol®   888   ATO   was   a   gift   of   Gattefossè   (Cedex,   France).   Stearic   acid   and   Tween®   80   were   purchased   from  Roig  Farma  (Barcelona,  Spain).  Phosphate  Buffer  Saline  (PBS)  was  provided   by  Sigma-­‐Aldrich  (Barcelona,  Spain).  Chloroform  and  ethyl  acetate  were  obtained   from   Panreac   Química   S.A.   (Barcelona,   Spain).   All   solvents   employed   for   the   chromatographic   analysis   were   of   analytical   grade.   Formic   acid   99   %   for   mass   spectroscopy   was   purchased   from   Fluka   (Barcelona,   Spain)   and   methanol   was   obtained  from  Merck  (Barcelona,  Spain).  All  other  chemicals  were  of  reagent  grade   and   used   without   further   purification.   Amicon   Ultra–15   centrifugal   filter   devices   were  purchased  from  Millipore  (Cork,  Ireland).   62  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

2.2.

Preparation  of  lipid  nanoparticles  

Lipid   nanoparticles   were   prepared   by   the   emulsification/solvent   evaporation   method.  For  the  simple  emulsion  solvent  evaporation  method,  edelfosine  and  the   lipid   (100   mg   of   either   Compritol®   or   stearic   acid)   were   dissolved   in   2   mL   of   chloroform.  This  solution  was  emulsified  with  10  mL  of  a  0.5  %  or  1  %  Tween  80   solution   by   ultrasonication   using   a   MicrosonTM   ultrasonic   cell   disruptor   (NY,   USA).   The  O/W  emulsion  formed  was  magnetically  stirred  for  45  minutes  and  subjected   to   low   vacuum   rotary   evaporation   for   the   complete   elimination   of   the   organic   solvent.   Particles   were   centrifuged   at   4500   g   for   40   minutes   using   an   Amicon   Ultra–15   filter   device   and   washed   twice   with   distilled   water.   The   obtained   particular   suspension   was   fast   frozen   under   –80   °C   for   at   least   3   hours   and   freeze-­‐ dried  in  order  to  store  it  at  4  °C.  For  the  double  emulsion  (W/O/W),  edelfosine  was   dissolved  in  100  μl  of  a  1  %  Tween  80  solution  and  emulsified  with  2  ml  of  ethyl   acetate.  This  first  W/O  emulsion  was  then  emulsified  with  10  ml  of  a  1  %  Tween   80   solution.   Following   steps   were   the   same   to   the   ones   of   the   simple   emulsion   solvent  evaporation  method.    

2.3.

Encapsulation  efficiency  

Edelfosine   was   extracted   by   dissolving   10   mg   of   nanoparticles   in   1   ml   of   chloroform   and   then   mixed   with   3   ml   of   ultra   pure   water.   The   mixture   was   vortexed   for   1   minute   and   then   centrifuged   at   9500   g   for   10   minutes.   The   supernatant   was   analysed   by   a   HPLC–MS   method,   which   is   a   slight   modification   of   a   previously   developed   method   [24].   The   apparatus   used   for   the   HPLC   analysis   was   a   Model   1100   series   LC   coupled   with   an   atmospheric   pressure   (AP)-­‐ electrospray   ionization   (ESI)   mass   spectrometer   (HP   1100   with   MSD   VL,   Waldbronn,   Germany).   Data   acquisition   and   analysis   were   performed   with   a   Hewlett-­‐Packard   computer   using   the   ChemStation   G2171   AA   programme.   Separation  was  carried  out  at  50  °C  on  a  reversed-­‐phase,  150  mm  

 3  mm  column  

packed   with   C18,   5   μm   silica   reversed-­‐phase   particles   (Gemini®)   obtained   from   Phenomenex®   (Torrance,   CA,   USA).   This   column   was   preceded   by   a   reversed-­‐ phase,  C18,  5  μm  guard  column  (SecurityGuard®,  20  mm  

 4  mm,  Phenomenex®,  

63  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

Torrance,  CA,  USA).  The  mobile  phase  was  a  mixture  of  methanol–1  %  formic  acid   (95:5,  v/v).  Separation  was  achieved  by  isocratic  solvent  elution  at  a  flow-­‐rate  of   0.5   ml/min.   The   mass   spectrometer   was   operated   in   the   positive   ESI   mode.   The   detection   of   edelfosine   was   performed   by   selected   ionization   monitoring   (SIM)   mode.  The  mass  spectrometer  was  programmed  to  monitor  the  ion  of  edelfosine  at   m/z  524.40.  Typical  retention  time  was  3.65  minutes.    

2.4.

Nanoparticle  characterization  

2.4.1. Particle  size,  size  distribution  and  zeta  potential   Particle   size   and   distribution   of   the   nanoparticles   were   measured   by   photon   correlation   spectroscopy   (PCS)   using   a   Zetasizer   Nano   (Malvern   Instruments,   UK).   Each   sample   was   diluted   30   fold   in   distilled   water   until   the   appropriate   concentration   of   particles   was   achieved   to   avoid   multiscattering   events.   The   obtained   homogenous   suspension   was   examined   to   determine   the   volume   mean   diameter,   size   distribution   and   polydispersity   and   repeated   three   times   for   each   sample.   The   data   are   expressed   as   a   mean   value   ±   standard   deviation.   Similarly,   the  zeta  potential  was  measured  using  the  same  equipment  described  previously   with   a   combination   of   laser   Doppler   velocimetry.   Samples   were   diluted   with   distilled  water  and  each  experiment  was  repeated  three  times.    

2.4.2. Morphology   Atomic   force   microscopy   (Cervantes   AFM   System,   Nanotec   Electrónica,   S.L.,   Spain)   was   employed   to   determine   the   shape   and   surface   morphology   of   the   nanoparticles.   AFM   was   conducted   with   Nanoscope   IIa   IIIa   in   the   tapping   mode.   The   nanoparticle   sample   was   mounted   on   a   metal   stab   and   scanned   by   the   AFM   maintained  in  a  constant  temperature  and  vibration  time  environment.      

2.4.3. Thermal  analysis  of  freeze-­‐dried  lipid  nanoparticles   The   thermal   characteristics   of   selected   batches   of   nanoparticles   were   determined  by  differential  scanning  calorimetry  thermal  analysis  using  a  2920  DSC   (Universal   V3.6C   TA   Instruments,   USA).   The   scan   rate   was   10   °C/min   in   the   64  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

temperature  range  from  –10  °C  to  275  °C  and  a  N2  flow  of  20  L/min.  An  empty  pan   was  used  as  reference  standard.  Indium  (purity  ≥99.95  %,  Fluka,  Switzerland)  was   employed  to  check  the  calibration  of  the  calorimetric  system.     2.4.4. X-­‐ray  studies  of  freeze-­‐dried  lipid  nanoparticles   X-­‐ray   diffraction   measurements   were   performed   in   order   to   clearly   elucidate   the   solid   state   of   both   lipids   and   drug   in   lipid   nanoparticles,   using   a   Bruker   D8   Advance  X-­‐ray  diffractometer  (Bruker  Biosciences  Española,  S.A.,  Spain).  The  X-­‐ray   diffractogram   was   scanned   with   the   diffraction   angle   increasing   from   2°   to   40°,   2θ   angle,  with  a  step  angle  of  0.02°  and  a  count  time  of  1  s  at  a  constant  temperature   of  25  °C.       2.5.

In  vitro  release  studies  

The   release   rate   of   edelfosine   from   lipid   nanoparticles   was   measured   in   PBS   medium  (pH  7.4).  Briefly,  5  mg  of  nanoparticles  were  dispersed  in  1  ml  of  buffer   solution   and   maintained   at   37   °C   under   stirring   (260   rpm).   At   appropriate   time   intervals,   samples   were   centrifuged   (23500   g,   10   minutes),   supernatants   were   filtrated  with  a  0.45  μm  pore  diameter  filter  and  kept  at  –20  °C  until  further  HPLC– MS  analysis  was  conducted  as  previously  described.  Three  samples  were  employed   for  each  time  and  the  study  was  performed  in  triplicate.   The   mean   in   vitro   dissolution   time   (MDT),   a   model-­‐independent   in   vitro   parameter   that   shows   the   meantime   for   edelfosine   to   release   from   the   lipid   nanoparticles   under   in   vitro   release   conditions,   was   calculated   according   to   the   equation:   Eq.  (1)     where  ABCin  vitro  is  the  area  between  the  release  curve  and  its  asymptote,  calculated   by  the  trapezoidal  rule  from  time  zero  to  the  last  measured  time  point,  and  M∞  is  

65  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

the  total  amount  of  released  drug  at  this  time  point.  The  release  rate  constant  (kd)   was  calculated  by  the  expression  kd  =  1/MDT.    

3. Results  and  discussion   3.1.

Particle  size,  size  distribution  and  zeta  potential  

Structurally,   edelfosine   has   a   structure   of   an   amphifile,   with   a   part   of   the   molecule  exhibiting  hydrophobicity  and  another  part  exhibiting  hydrophilicity,  like   a  surface–active  agent  (Fig.1).    

  Figure  1.  Chemical  structure  of  edelfosine      

Edelfosine-­‐loaded   lipid   nanoparticles   were   obtained   by   either   simple   or   multiple   emulsion   solvent   evaporation   method,   and   freeze-­‐dried.   These   nanoparticles   were   then   characterized   to   assess   the   effect   of   the   different   lipids   and  surfactant  concentrations  on  mean  particle  size,  size  distribution  and  surface   charge.   Using   lipids   as   matrices   for   the   particles,   different   characteristics   can   be   obtained   by   optimizing   the   formulation   parameters   such   as   type   of   lipids,   surfactants,  organic  solvents  and  emulsifying  procedure  chosen  [18].  The  presence   of  a  non-­‐ionic  surfactant  is  important  to  reduce  the  dynamic  interfacial  tension  and   to   stabilize   the   nanosuspension.   The   surfactant   is   adsorbed   on   the   nanoparticle   surface,  increasing  the  steric  repulsion  between  particles.  In  this  study,  Tween®  80   was   tested   at   two   different   concentrations  (w/v),  being  sufficient  to  obtain  small   lipid  nanoparticles  and  permitting  the  removal  of  its  excess  by  centrifugation  and   washing.  

66  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

The   average   size   and   polydispersity   indices   of   lipid   nanoparticles   formulated   with  different  lipids  are  reported  in  Table  1.  All  data  are  expressed  as  mean  value  ±   standard  deviation.      

Table  1.  Average  size,  polydispersity  index  (PDI)  and  zeta  potential  of  lipid  nanoparticles  prepared   either  with  methylene  chloride  (MC)  or  trichloromethane  (TCM).   Lipid

Solvent

Stabilizing agent

Size (nm)

PDI

ζ Potential (mV)

Stearic acid

MC

0.5% Tween 80

TCM Compritol® 888 ATO

TCM

480 ± 10.79

0.204 ± 0.01

–32,75

1% Tween 80

611 ± 5.29

0.626 ± 0.13

–35,15

0.5% Tween 80

415 ± 30.09

0.451 ± 0.09

–29,33

1% Tween 80

312 ± 7.51

0.225 ± 0.04

–32,4

1% Tween 80

372 ± 18.84

0.354 ± 0.06

–32,3

 

 

For  all  lipids,  it  was  possible  to  obtain  submicron  sized  lipid  nanoparticles  with   two   different   concentrations   of   Tween®   80.   In   fact,   all   edelfosine   loaded   formulations  showed  a  mean  diameter  in  the  range  of  300  –  600  nm.  There  was  a   decrease   in   size   of   particles   when   formulated   with   edelfosine   (data   not   shown),   probably   due   to   the   effect   of   edelfosine   as   a   surfactant   agent.   Smallest   particles   were  obtained  using  stearic  acid,  chloroform  and  an  aqueous  solution  of  Tween®   80  at  a  concentration  of  1  %.  The  polydispersity  index  (PDI)  value  was  in  the  range   of  0.2  –  0.6  for  all  lipid  nanoparticles  investigated.     Zeta  potential  can  make  a  prediction  about  the  stability  of  colloid  dispersions.  A   high   zeta   potential   (>   30   mV)   can   provide   an   electric   repulsion   to   avoid   the   aggregation   of   particles   [25].   The   incorporation   of   edelfosine   into   lipid   nanoparticles  had  no  significant  influence  on  the  zeta  potentials  of  particles,  which   was   negative   in   all   cases.   However,   as   the   concentration   of   surfactant   was   increased   in   the   formulation   the   zeta   potential   was   found   to   be   more   negative   (Table   1).   This   could   be   due   to   the   presence   of   oleic   acid   traces   in   Tween   on   the   particle   surface,   forming   a   denser   surfactant   film,   thus   eliciting   an   increased   electrophoretic  mobility.        

67  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

3.2.

Drug  entrapment  efficiency  and  loading  capacity  

Over   the   past   few   years   many   different   drugs   had   been   successfully   incorporated   in   lipid   nano–   or   microparticles   [20,   26-­‐28].   Relatively   high   drug   encapsulation  efficiency  for  hydrophobic  drugs  was  one  of  the  major  advantages  of   lipid   nanoparticles   [20].   It   is   also   known   that   the   lipid   crystalline   structure   related   to   the   chemical   nature   of   the   lipid   is   a   key   factor   to   determine   whether   a   drug   will   be   expelled   or   firmly   incorporated   into   the   carrier   systems.   In   the   nanoparticle   structure,  the  lipid  forming  highly  crystalline  state  with  a  perfect  lattice  would  lead   to   drug   expulsion.   On   the   other   hand,   imperfections   (lattice   defects)   of   the   lipid   structure   could   offer   space   to   accommodate   the   drugs   [29].   As   a   result,   the   structure  of  less  ordered  arrangement  in  the  nanoparticles  would  be  beneficial  to   the  drug  loading  capacity  like  the  samples  in  this  study.      

Table  2.  Encapsulation  efficiency  (EE)  of  the  different  nanoparticles  prepared  either  with   methylene  chloride  (MC)  or  trichloromethane  (TCM).  

Lipid

Solvent

Stabilizing agent

%EE

Stearic acid

MC

Tween 80 0,5%

5,10

Tween 80 1%

4,00

Tween 80 0,5%

9,20

Tween 80 1%

6,40

Tween 80 1%

84,35

TCM Compritol® 888 ATO

TCM

 

 

From   the   results   listed   in   Table   2,   it   can   be   observed   that   the   entrapment   efficiency  of  edelfosine  in  the  lipid  nanoparticles  prepared  by  the  simple  emulsion   formation   solvent   evaporation   method   ranged   from   about   4   to   10   %   for   stearic   acid  nanoparticles.  On  the  other  hand,  nanoparticles  formulated  using  Compritol®   encapsulated   more   than   80   %   of   the   drug   (Table   2).   This   high   encapsulation   efficiency  in  comparison  to  the  stearic  acid  nanoparticles  is  likely  to  be  due  to  the   partially  amorphous  state  of  the  Compritol®  in  the  formulation,  which  allows  more   edelfosine  to  be  incorporated  among  lipid  chains.  It  can  also  be  observed  that  the   emulsifier  concentrations  investigated  in  this  study  (0.5  %  and  1  %  Tween  80)  do   not  affect  the  encapsulation  efficiency.  

68  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

On   the   other   hand,   lipid   nanoparticles   prepared   by   the   multiple   emulsion   solvent   evaporation   method   showed   very   low   encapsulation   efficiency,   less   than   1   %  (data  not  shown).     3.3.

Morphology  

In   order   to   investigate   the   shape   and   surface   morphology   of   the   Compritol®   nanoparticles,   atomic   force   microscopy   was   employed.   The   AFM   images   reveal   the   fine   structure   of   the   Compritol®   lipid   nanoparticle   surface   (Fig.   2A).   They   give   clear   3D   morphological   images   of   spherical   nanoparticles   of   sub–400   nm   diameter   and   they   also   confirm   that   there   was   no   aggregation   or   adhesion   among   the   nanoparticles  (Fig.  2C).  Furthermore,  the  surface  morphology  of  the  nanoparticles   could   be   seen   closely   from   the   AFM   images.   It   was   noticeable   from   the   zoom–in   picture  (Fig.  2B)  the  smooth  surface  morphology  of  the  nanoparticles.      

A  

B  

           

C  

        Figure   2.   Atomic   force   microscopy   images   of   freeze-­‐dried   Compritol®   lipid   nanoparticles:   multi-­‐ particles  (A);  zoom-­‐in  of  the  selected  area  of  A  (B);  3D  morphological  image  (C).    

69  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

3.4.

DSC  and  X-­‐ray  diffractometry  assays  

Lipid   nanoparticles   were   analysed   by   DSC   and   X-­‐ray   diffractometry   to   investigate   the   crystal   pattern   of   both   edelfosine   and   lipids,   because   this   aspect   could   influence   the   in   vitro   and   in   vivo   release   of   the   drug   from   the   systems.   To   probe   this   effect,   analysis   were   performed   on   the   following   samples:   edelfosine;   Compritol®   888   ATO;   stearic   acid;   edelfosine   loaded   stearic   acid   nanoparticles;   edelfosine   loaded   Compritol®   nanoparticles;   unloaded   stearic   acid   nanoparticles;   unloaded  Compritol®  nanoparticles.  

  Figure   3.   DSC   thermograms   of:     (a)   edelfosine,   (b)   stearic   acid,   (c)   unloaded   stearic   acid   lipid   nanoparticles  and  (d)  edelfosine-­‐loaded  stearic  acid  lipid  nanoparticles  (Fig.  3A);  (a)  edelfosine,  (b)   Compritol®,  (c)  unloaded  Compritol®  lipid  nanoparticles  and  (d)  edelfosine-­‐loaded  Compritol®  lipid   nanoparticles  (Fig.  3B).  

70  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

Fig.   3   depicts   the   DSC   thermograms   of   edelfosine   loaded   and   unloaded   lyophilised  nanoparticles.  As  Compritol®  is  not  composed  of  pure  triacylglycerols,   the   observed   melting   peak   at   72.43   °C   might   be   due   to   a   mixture   of   metastable   polymorphic  β  and  β’  forms.  The  heating  run  showed  the  just  mentioned  melting   event  at  72.43  °C  and  a  relatively  small  endothermic  shoulder  at  around  51  °C.  This   small   shoulder   corresponds   to   the   melting   of   a   very   unstable   modification   of   Compritol®,   which   is   the   α   modification   [30],   that   clearly   disappears   after   the   treatment  with  organic  solvents  for  the  nanoparticle  preparation.  DSC  analysis  of   edelfosine-­‐loaded  lipid  nanoparticles  showed  that  the  drug  melting  peak  at  223°C   is  present  neither  in  the  stearic  acid  lipid  nanoparticles  nor  in  the  Compritol®  lipid   nanoparticles   whereas   for   the   pure   drug,   the   melting   peak   occurs   before   its   decomposition.   This   thermal   behaviour   may   be   ascribed   to   the   presence   of   edelfosine   in   an   amorphous   form   or   molecularly   dispersed,   but   it   may   also   be   related   to   a   possible   solubilisation   of   the   drug   in   the   molten   lipid   when   the   DSC   assay   was   performed.   Besides,   X-­‐ray   diffraction   studies   support   this   theory   showing  a  partial  crystalline  state  of  the  drug.  This  effect  on  the  crystalline  habit  of   edelfosine  may  be  related  to  the  preparative  method  of  the  lipid  nanoparticles,  in   which  edelfosine  may  be  turned  from  a  crystalline  state  to  an  amorphous  one  by   the  use  of  organic  solvents  like  chloroform  or  methylene  chloride.    Thermal   behaviour   of   lipids   can   also   explain   the   different   encapsulation   efficiency   of   edelfosine.   Unloaded   stearic   acid   nanoparticles   showed   the   melting   peak   of   the   stearic   acid   at   59   °C,   indicating   its   presence   in   crystalline   state,   thus   letting   less   amount   of   drug   to   be   incorporated   among   its   lipidic   chains.   On   the   other   hand,   Compritol®   nanoparticles   seem   to   lose   part   of   their   crystalline   state   transforming   from   a   mixture   of   β   and   β’   polymorphs   to   the   most   stable   β   polymorph,   permitting   edelfosine   to   fit   in   the   molecular   gaps.   These   findings   were   confirmed  by  X-­‐ray  diffractometry  assays.          

71  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

                                    Figure   4.   X-­‐ray   diffractograms   of:   (a)   edelfosine,   (b)   stearic   acid,   (c)   unloaded   stearic   acid   lipid   nanoparticles   and   (d)   edelfosine-­‐loaded   stearic   acid   lipid   nanoparticles   (A);   (a)   edelfosine,   (b)   Compritol®,  (c)  edelfosine-­‐loaded  Compritol®  lipid  nanoparticles  and  (d)  unloaded  Compritol®  lipid   nanoparticles  (B).  

  Unloaded   stearic   acid   nanoparticles   (Fig.   4A)   show   two   sharp   peaks   corresponding  to  those  of  the  stearic  acid,  whereas  the  diffraction  pattern  of  bulk   Compritol®   (Fig.   4B)   showed   two   main   typical   signals   at   21.5   (2θ)   and   23.5   (2θ)   that  are  significantly  modified  when  formulated  into  nanoparticles.  Besides,  when   Compritol®   nanoparticles   are   formulated,   another   signal   arises   at   19.4   (2θ)   (Fig.   4B)  which  corresponds  to  the  most  stable  polymorphic  form  of  triacylglycerols  β   [30].   These   results   might   indicate   that   the   final   formulation   is   composed   of   the   72  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

most   stable   polymorphic   state   of   Compritol®.   Comparing   these   results   with   edelfosine   loaded   lipid   nanoparticles,   some   crystalline   drug   signal   could   still   be   detected,   indicating   a   possible   coexistence   of   edelfosine   in   both   crystalline   and   amorphous   states,   being   this   last   one   the   predominant.   As   a   result,   edelfosine   would   enrich   the   particle   surface   when   formulated   with   stearic   acid,   whereas   it   would  be  incorporated  among  the  lipid  chains  of  Compritol®.       3.5.

In  vitro  release  studies  

The  amount  of  edelfosine  released  from  lipid  nanoparticles  was  determined  by   an   in   vitro   release   assay,   in   an   effort   to   assess   whether   edelfosine–incorporating   lipid  nanoparticles  might  be  useful  as  a  sustained-­‐release  dosage  form.   Fig.  5  displays  the  release  profiles  for  nanoparticles  fabricated  with  stearic  acid   or   Compritol®   using   1   %   Tween   80   as   emulsifier.   When   edelfosine   was   incorporated   in   stearic   acid   nanoparticles,   a   63   %   burst   release   of   the   drug   was   observed   within   20   minutes   (Fig.5).   Conversely,   the   initial   release   burst   for   nanoparticles   prepared   using   Compritol®   was   less   than   40   %   within   the   first   20   minutes.                   Figure   5.   In   vitro   release   profiles   of   edelfosine   from   stearic   acid   lipid   nanoparticles   (■) and   Compritol®  888  ATO  nanoparticles  (▲).  

The   reason   for   the   high   initial   release   of   edelfosine   from   the   stearic   acid   nanoparticles   could   be   the   diffusion   release   of   edelfosine   distributed   near   the  

73  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

surface   and   in   the   outer   portion   of   the   nanoparticles   [12].   These   results   were   in   accordance  with  the  observations  made  by  DSC  and  X-­‐ray  diffractometry.   Afterwards,  the  release  rate  slowed  for  both  formulations,  reaching  55  %  after   24   h   for   nanoparticles   prepared   with   Compritol®,   and   98   %   for   the   stearic   acid   nanoparticles.     The  release  of  a  drug  from  the  dosage  form  implies  a  crucial  step,  which  is  the   dissolution   of   the   drug.   This   process   is   ruled   by   a   release   rate   constant   (kd)   that   can  be  easily  estimated  and  it  is  different  depending,  among  other  factors,  on  the   composition   of   the   particle.   In   our   study,   this   constant   was   estimated   for   the   release   of   edelfosine   from   lipid   nanoparticles   formulated   either   with   stearic   acid   or  Compritol®.   Results  showed  that  there  is  a  different  release  profile  depending  on  the  lipid.   Stearic   acid   presents   a   higher   release   rate   constant   (kd   =   0.208)   than   Compritol®   (kd  =  0.178),  indicative  of  a  faster  release.  Besides,  there  seems  to  be  no  influence   of  the  amounts  of  surfactant  employed  in  the  release  kinetics  of  the  drug,  since  the   release   rate   constant   from   nanoparticles   prepared   with   different   surfactant   concentrations  (0.5  or  1  %  Tween  80)  were  equivalent.   Nevertheless,  the  in  vitro  release  rates  cannot  be  directly  extrapolated  to  predict   behaviour  of  these  systems  in  biological  environments.  The  differences  between  in   vitro  and  in  vivo  degradation  of  the  nanoparticles  can  be  ascribed  to  the  presence   of   enzymes   [31]   and   the   prolonged   release   of   the   drug   from   the   nanoparticulate   systems  has  to  be  quantified  in  an  in  vivo  animal  model.    

4. Conclusions   The   present   research   paper   proposed   a   novel   formulation   for   edelfosine   by   using  lipid  nanoparticles.  It  can  be  concluded  that  Compritol®  presents  advantages   as   a   matrix   material   for   the   manufacture   of   the   nanoparticles   and   the   controlled   release  of  edelfosine.   Current   studies   are   aimed   at   evaluating   the   in   vitro   (in   cell   lines)   and   in   vivo   efficacy  of  these  newly  developed  formulations.  

74  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

 

Acknowledgements   This  work  was  supported  by  “Fundación  Caja  Navarra”.  The  authors  are  grateful  to   Nanotec   Electrónica   S.L.   and   Dr.   Blanca   Galar   for   her   assistance   in   X–Ray   diffraction   studies.   The   first   author   acknowledges   a   fellowship   from   the   Department  of  Education  of  the  Basque  Government  (BFI06.37).    

75  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

References     1.  

Gajate,   C.   and   F.   Mollinedo,   Biological   activities,   mechanisms   of   action   and   biomedical   prospect   of   the   antitumor   ether   phospholipid   ET-­‐18-­‐OCH3   (edelfosine),   a   proapoptotic   agent   in   tumor   cells.   Curr.   Drug.   Metab.,   2002.   3(5):  p.  491-­‐525.  

2.  

Berdel,  W.E.,  U.  Fink,  and  J.  Rastetter,  Clinical  phase  I  pilot  study  of  the  alkyl   lysophospholipid  derivative  ET-­‐18-­‐OCH3.  Lipids,  1987.  22(11):  p.  967-­‐9.  

3.  

Mollinedo,   F.,   et   al.,   Selective   induction   of   apoptosis   in   cancer   cells   by   the   ether   lipid   ET-­‐18-­‐OCH3   (Edelfosine):   molecular   structure   requirements,   cellular   uptake,   and   protection   by   Bcl-­‐2   and   Bcl-­‐X(L).   Cancer   Res.,   1997.   57(7):  p.  1320-­‐8.  

4.  

Gajate,   C.   and   F.   Mollinedo,   Edelfosine   and   perifosine   induce   selective   apoptosis   in   multiple   myeloma   by   recruitment   of   death   receptors   and   downstream  signaling  molecules  into  lipid  rafts.  Blood,  2007.  109(2):  p.  711-­‐ 9.  

5.  

Mollinedo,   F.,   Antitumour   ether   lipids:   proapoptotic   agents   with   multiple   therapeutic  indications.  Expert  Opin.  Therap.  Patents.,  2007.  17(4):  p.  385-­‐ 405.  

6.  

Azzouz,   S.,   et   al.,   Leishmanicidal   activity   of   edelfosine,   miltefosine   and   ilmofosine.  Basic  Clin  Pharmacol  Toxicol,  2005.  96(1):  p.  60-­‐5.  

7.  

Boggs,   K.P.,   C.O.   Rock,   and   S.   Jackowski,   Lysophosphatidylcholine   and   1-­‐O-­‐ octadecyl-­‐2-­‐O-­‐methyl-­‐rac-­‐glycero-­‐3-­‐phosphocholine   inhibit   the   CDP-­‐choline   pathway   of   phosphatidylcholine   synthesis   at   the   CTP:phosphocholine   cytidylyltransferase  step.  J.  Biol.  Chem.,  1995.  270(13):  p.  7757-­‐64.  

8.  

Vogler,   W.R.,   et   al.,   The   anticancer   drug   edelfosine   is   a   potent   inhibitor   of   neovascularization  in  vivo.  Cancer  Invest,  1998.  16(8):  p.  549-­‐53.  

76  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

9.  

Zheng,   B.,   et   al.,   Inhibition   of   protein   kinase   C,   (sodium   plus   potassium)-­‐ activated   adenosine   triphosphatase,   and   sodium   pump   by   synthetic   phospholipid  analogues.  Cancer  Res,  1990.  50(10):  p.  3025-­‐31.  

10.  

Ahmad,   I.,   et   al.,   Enhanced   therapeutic   effects   of   liposome-­‐associated   1-­‐O-­‐ octadecyl-­‐2-­‐O-­‐methyl-­‐sn-­‐glycero-­‐3-­‐phosphocholine.  

Cancer  

Res.,  

1997.  

57(10):  p.  1915-­‐21.   11.  

Bhamra,   R.,   et   al.,   Activity,   pharmacokinetics   and   tissue   distribution   of   TLC   ELL-­‐12   (liposomal   antitumor   ether   lipid)   in   rats   with   transplantable,   s.c.   methylnitrosourea-­‐induced  tumors.  Anticancer  Drugs,  2003.  14(6):  p.  481-­‐6.  

12.  

zur  Muhlen,  A.,  C.  Schwarz,  and  W.  Mehnert,  Solid  lipid  nanoparticles  (SLN)   for   controlled   drug   delivery-­‐-­‐drug   release   and   release   mechanism.   Eur.   J.   Pharm.  Biopharm.,  1998.  45(2):  p.  149-­‐55.  

13.  

Siekmann,   B.   and   K.   Westesen,   Sub-­‐micron   sized   parenteral   carrier   systems   based  on  solid  lipids.  Pharm.  Pharmacol.  Lett.,  1992.  1:  p.  123.  

14.  

Schwarz,   C.   and   W.   Mehnert,   Solid   lipid   nanoparticles   (SLN)   for   controlled   drug   delivery.   II.   Drug   incorporation   and   physicochemical   characterization.   J.   Microencapsul.,  1999.  16(2):  p.  205-­‐13.  

15.  

de   Vringer,   T.   and   H.A.   de   Ronde,   Preparation   and   structure   of   a   water-­‐in-­‐oil   cream  containing  lipid  nanoparticles.  J.  Pharm.  Sci.,  1995.  84(4):  p.  466-­‐72.  

16.  

Almeida,   A.J.,   S.   Runge,   and   R.H.   Muller,   Peptide-­‐loaded   solid   lipid   nanoparticles  (SLN):  Influence  of  production  parameters.  Int.  J.  Pharm.,  1997.   149(2):  p.  255-­‐265.  

17.  

Wissing,   S.A.,   O.   Kayser,   and   R.H.   Muller,   Solid   lipid   nanoparticles   for   parenteral  drug  delivery.  Adv.  Drug  Deliv.  Rev.,  2004.  56(9):  p.  1257-­‐72.  

18.  

Mehnert,   W.   and   K.   Mader,   Solid   lipid   nanoparticles:   production,   characterization   and   applications.   Adv.   Drug   Deliv.   Rev.,   2001.   47(2-­‐3):   p.   165-­‐96.  

77  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

19.  

Hu,   F.Q.,   et   al.,   Preparation   and   characterization   of   stearic   acid   nanostructured   lipid   carriers   by   solvent   diffusion   method   in   an   aqueous   system.  Colloids  Surf.  B  Biointerfaces.,  2005.  45(3-­‐4):  p.  167-­‐73.  

20.  

Jaspart,   S.,   et   al.,   Solid   lipid   microparticles   as   a   sustained   release   system   for   pulmonary  drug  delivery.  Eur.  J.  Pharm.  Biopharm.,  2007.  65(1):  p.  47-­‐56.  

21.  

Muller,   R.H.,   M.   Radtke,   and   S.A.   Wissing,   Nanostructured   lipid   matrices   for   improved  microencapsulation  of  drugs.  Int.  J.  Pharm.,  2002.  242(1-­‐2):  p.  121-­‐ 8.  

22.  

Freitas,  C.  and  R.H.  Muller,  Effect  of  light  and  temperature  on  zeta  potential   and   physical   stability   in   solid   lipid   nanoparticles   (SLN)   dispersion.   Int.   J.   Pharm.,  1998.  168:  p.  221-­‐229.  

23.  

Cavalli,  R.,  et  al.,  Sterilization  and  freeze  -­‐drying  of  drug-­‐free  and  drug-­‐loaded   solid  lipid  nanoparticles.  Int.  J.  Pharm.,  1997.  148:  p.  47-­‐54.  

24.  

Blanco-­‐Prieto,   M.J.,   M.A.   Campanero,   and   F.   Mollinedo,   Quantitative   determination   of   the   antitumor   alkyl   ether   phospholipid   edelfosine   by   reversed-­‐phase   liquid   chromatography-­‐electrospray   mass   spectrometry:   application   to   cell   uptake   studies   and   characterization   of   drug   delivery   systems.  J.  Chromatogr.  B.  Analyt.  Technol.  Biomed.  Life  Sci.,  2004.  810(1):   p.  85-­‐92.  

25.  

Levy,  M.Y.,  et  al.,  Characterization  of  diazepam  submicron  emulsion  interface:   role  of  oleic  acid.  J.  Microencapsul.,  1994.  11(1):  p.  79-­‐92.  

26.  

Chen,  H.,  et  al.,  Podophyllotoxin-­‐loaded  solid  lipid  nanoparticles  for  epidermal   targeting.  J.  Control.  Rel.,  2006.  110(2):  p.  296-­‐306.  

27.  

Hu,   F.Q.,   et   al.,   Preparation   of   solid   lipid   nanoparticles   with   clobetasol   propionate   by   a   novel   solvent   diffusion   method   in   aqueous   system   and   physicochemical  characterization.  Int.  J.  Pharm.,  2002.  239(1-­‐2):  p.  121-­‐8.  

28.  

Lee,   M.K.,   S.J.   Lim,   and   C.K.   Kim,   Preparation,   characterization   and   in   vitro   cytotoxicity  of  paclitaxel-­‐loaded  sterically  stabilized  solid  lipid  nanoparticles.   Biomaterials,  2007.  28(12):  p.  2137-­‐46.  

78  

Chapter  2.  Lipid  nanoparticles  for  alkyl  lysophospholipid  edelfosine  encapsulation:   development  and  in  vitro  characterization    

29.  

Hou,  D.,  et  al.,  The  production  and  characteristics  of  solid  lipid  nanoparticles   (SLNs).  Biomaterials,  2003.  24(10):  p.  1781-­‐5.  

30.  

Souto,   E.B.,   W.   Mehnert,   and   R.H.   Muller,   Polymorphic   behaviour   of   Compritol888  ATO  as  bulk  lipid  and  as  SLN  and  NLC.  J.  Microencapsul.,  2006.   23(4):  p.  417-­‐33.  

31.  

Olbrich,  C.  and  R.H.  Muller,  Enzymatic  degradation  of  SLN-­‐effect  of  surfactant   and  surfactant  mixtures.  Int.  J.  Pharm.,  1999.  180(1):  p.  31-­‐9.  

                         

79  

 

   

   

 

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL      

   

Chapter  3   Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid   edelfosine:  Pharmacokinetic  profile,  biodistribution   studies  and  in  vivo  efficacy  against  mantle-­‐cell  lymphoma   Ander  Estella-­‐Hermoso  de  Mendoza1,  Miguel  A.  Campanero2,  Hugo  Lana1,  Janny  A.   Villa-­‐Pulgarin3,4,  Janis  de  la  Iglesia-­‐Vicente3,  Faustino  Mollinedo3,  María  J.  Blanco-­‐ Prieto1    

1Departamento  de  Farmacia  y  Tecnología  Farmacéutica,  Facultad  de  Farmacia,  

University  of  Navarra,  E-­‐31008,  Spain   2Servicio  de  Farmacología  Clínica,  Clínica  Universitaria,  E-­‐31080  Pamplona,  Spain   3Centro  de  Investigación  del  Cáncer,  Instituto  de  Biología  Molecular  y  Celular  del  

Cáncer,  CSIC-­‐Universidad  de  Salamanca,  Campus  Miguel  de  Unamuno,  E-­‐37007   Salamanca,  Spain   4APOINTECH,  Centro  Hispano-­‐Luso  de  Investigaciones  Agrarias  (CIALE),  Parque  

Científico  de  la  Universidad  de  Salamanca,  C/  Rio  Duero  12,  E-­‐37185  Villamayor,   Salamanca,  Spain  

 

Key   Words:   edelfosine,   lipid   nanoparticles,   bioavailability,   pharmacokinetics,   biodistribution,  lymphoma   Corresponding   author:   Dr.   María   J.   Blanco-­‐Prieto,   Department   of   Pharmacy   and   Pharmaceutical   Technology,   School   of   Pharmacy,   University   of   Navarra,   C/Irunlarrea   1,   E-­‐31080   Pamplona,   Spain,   Office   phone:   +   34   948   425   600   ext.   6519,  Fax:  +  34  948  425  649,  e-­‐mail:  [email protected]    

 

Submitted  for  evaluation.  

81  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

  Abstract     To   increase   the   oral   bioavailability   of   the   antitumor   drug   edelfosine,   lipid   nanoparticles   (LN)   made   of   synthetic   lipids   Compritol®   888   ATO   and   Precirol®   ATO  5  were  developed.  Average  size  of  particles  was  110.4  ±  2.1  nm  and  103.1  ±   2.9   nm,   for   Compritol®   and   Precirol®   drug-­‐loaded   LN.   Zeta   potential   was   close   to   -­‐ 21   mV   in   all   cases,   and   encapsulation   efficiency   of   edelfosine   reached   85   %   for   both  type  of  lipids.  Pharmacokinetic  and  biodistribution  profiles  of  the  drug  were   studied   after   intravenous   and   oral   administration   of   edelfosine-­‐containing   LN.   Results  showed  that  encapsulation  of  edelfosine  in  LN  decreases  hemolytic  toxicity   of  the  drug  in  90  %  and  provides  an  increase  in  relative  oral  bioavailability  of  1500   %   after   a   single   oral   administration   of   drug-­‐loaded   LN,   maintaining   edelfosine   plasma  levels  over  7  days  in  contrast  to  a  single  oral  administration  of  edelfosine   solution,   which   presents   a   relative   oral   bioavailability   of   10   %.   Pharmacokinetic   parameters   confirmed   the   slow   elimination   of   the   drug   and   a   wide   distribution   throughout  the  body.  Moreover,  edelfosine-­‐loaded  LN  showed  a  high  accumulation   of  the  drug  in  lymph  nodes  and  showed  slower  tumor  growth  than  the  free  drug  in   a  murine  lymphoma  xenograft  model,  as  well  as  potent  extranodal  dissemination   inhibition.    

83  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

1. Introduction   Edelfosine   is   considered   the   prototype   of   a   promising   class   of   antitumor   agents,   collectively  known  as  alkyl-­‐lysophospholipid  analogues  or  antitumor  ether  lipids.   These   agents   present   the   singular   characteristic   of   not   targeting   the   DNA,   but   affecting   the   cell   membrane   and   the   apoptotic   machinery   of   the   cancer   cell   [1].   Recent   in   vitro   studies   have   shown   that   edelfosine   is   preferentially   uptaken   by   tumoral  cells,  sparing  normal  cells  [2].     However,   edelfosine   presents   some   drawbacks   when   administered   intravenously,  as  dose-­‐dependent  hemolysis  hampers  its  administration  at  certain   doses  [3].  In  addition,  edelfosine  presents  bioavailability  values  below  10  %  after  a   single  oral  administration  of  30  mg/kg;  however,  this  bioavailability  increased  to   64  %  after  multiple  oral  administration  of  the  same  dose  after  six  days  [2].     Owing  to  the  drawbacks  of  this  molecule,  there  has  been  an  attempt  to  design   new   drug   delivery   systems   that   can   modify   the   absorption   rate,   selectively   transport   the   drug   to   the   target,   modifying   the   drug   distribution   profile   and   extending   the   drug   release   time   in   order   to   improve   drug   bioavailability,   and   decrease  its  toxicity.  Among  the  different  lipid-­‐made  colloidal  carriers,  edelfosine   was   incorporated   into   liposomes   [4]   and   lipid   nanoparticles   (LN)   made   of   biocompatible   lipids   [5].   The   liposomal   formulation   was   able   to   prevent   the   hemolytic   toxicity   of   the   drug,   but   the   main   inconvenience   found   was   its   rapid   clearance   from   plasma.   Edelfosine-­‐loaded   liposomes   showed   both   in   vivo   and   in   vitro  activity  against  methylnitrosourea-­‐induced  tumors,  and  it  was  approximately   4   -­‐   8   times   less   acutely   toxic   than   free   edelfosine.   After   a   slow   intravenous   (i.v.)   bolus   injection   of   12.5   mg/kg   of   edelfosine   delivered   as   edelfosine-­‐loaded   liposomes   to   rats,   several   parameters   were   determined.   Terminal   half-­‐life   (t1/2)   value  was  approximately  13.1  h.  Clearance  (CL)  and  volume  of  distribution  at  the   steady-­‐state   (Vss)   values   were   estimated   at   0.0161   L/h/kg   and   0.203   L/kg,   respectively   [4].   Edelfosine-­‐loaded   LN   developed   by   our   group   were   considered   another   alternative   to   deliver   the   drug   to   the   organism   [5].   These   carriers   are   colloidal   transporters   composed   of   a   biocompatible   and   biodegradable   lipid   matrix.   They   combine   advantages   of   liposomes,   polymeric   nanoparticles   and   85  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

emulsions,  while  diminishing  possible  drawbacks  associated  with  them  [6].  Lipids   employed   to   form   these   lipid   cores   are   biodegradable   raw   materials   that   are   biocompatible:  triglycerides  (i.e.  tristearin),  partial  glycerides  (i.e.  Compritol®  888   ATO  and  Precirol®  ATO  5),  fatty  acids  (i.e.  stearic  acid),  steroids  (i.e.  cholesterol)  or   waxes   (i.e.   cetyl   palmitate)   [7].   The   formulation   methods   are   also   diverse   [7,   8].   However,   most   techniques   employ   organic   solvents,   which   may   imply   regulatory   and   toxicity   issues.   Moreover,   an   improvement   of   LN   over   the   liposomes   is   their   physical   and   chemical   long-­‐term   stability   up   to   12   -­‐   24   months   [9].   The   freeze-­‐ drying   process   of   LN   has   been   shown   to   increase   their   physicochemical   stability   over   long   periods   of   time   [10].   LN   may   also   be   used   to   target   drugs   at   particular   organs   without   surface   modification   because   of   their   small   size   (50   -­‐   200   nm).   Besides,   LN   have   attracted   rising   interest   for   their   ability   to   overcome   certain   biological  barriers,  resulting  in  increased  therapeutic  efficacy  of  the  encapsulated   drug  and  increase  in  tumor  accumulation  [11].     Mantle  cell  lymphoma  (MCL)  is  a  B-­‐cell  malignancy  that  comprises  about  7  %  of   all   non-­‐Hodgkin’s   lymphomas   (NHLs),   which   is   characterized   clinically   by   extranodal  disease  in  older  male  patients  who  present  at  an  advanced  stage  [12].   Even   though   most   patients   initially   gain   a   benefit   from   systemic   treatments,   the   responses  obtained  are  generally  of  limited  duration.  As  a  result,  patients  generally   relapse   with   less   responsive   disease,   showing   a   consistently   bad   outcome   with   a   median  overall  survival  from  diagnosis  of  43  months  [13,  14].  Several  approaches   using   more   severe   combination   chemotherapy,   namely   stem   cell   transplantation,   have  shown  higher  response  rates  and  more  lasting  remission  in  selected  patients,   but   the   greater   part   of   MCL   patients   are   not   candidates   for   such   dose-­‐intensive   regimens   [15,   16].   MCL   has   recently   become   an   area   of   intense   clinical   research,   and   it   appears   that   median   overall   survival   may   be   improving,   but   MCL   is   still   considered   incurable   with   current   treatments   [17].   Although   several   cytotoxic   combinations   including   cisplatin,   gemcitabine,   carmustine   and   other   alkylating   drugs   have   been   employed,   their   inherent   toxicity   is   considered   the   main   drawback  at  the  time  of  election  [18].    

86  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

In   a   previous   study,   we   determined   that   edelfosine   displays   a   biexponential   pharmacokinetic  behavior  in  mice,  presenting  no  significant  differences  regardless   of   the   mouse   strain   employed   [2].   The   tissue   distribution   of   edelfosine   in   mice   shows   that   the   drug   is   widely   scattered   across   different   organs,   although   it   is   preferentially  internalized  by  the  tumor  both  in  vitro  and  in  vivo.  The  present  work   tries   to   point   out   how   the   biodistribution   and   pharmacokinetic   profile   of   edelfosine  is  altered  compared  to  that  of  the  free  drug,  when  it  is  encapsulated  in   LN.  The  efficacy  of  the  chemotherapeutic  potential  of  edelfosine  loaded  LN  via  the   oral  route  in  experimental  murine  lymphoma  xenograft  model  was  also  evaluated.    

2. Materials  and  Methods   2.1.

Chemicals  

Edelfosine  was  from  APOINTECH  (Salamanca,  Spain).  Compritol®  888  ATO  and   Precirol®   ATO   5   were   a   gift   from   Gattefossé   (Lyon,   France).   Tween®   80   was   obtained   from   Roig   Farma   (Barcelona,   Spain).   Platelet   Activating   Factor   (PAF)   and   PBS   (10   mM   phosphate,   0.9   %   NaCl)   were   obtained   from   Sigma-­‐Aldrich   (Madrid,   Spain).   Chloroform   was   purchased   from   Panreac   (Madrid,   Spain)   and   methanol   was  obtained  from  Merck  (Barcelona,  Spain).  All  other  solvents  were  of  analytical   grade.     2.2.

Cell  culture  

The  human  mantle-­‐cell  lymphoma  cell  line  JVM-­‐2  (DSMZ,  Germany)  was  grown   in   RPMI-­‐1640   containing   10   %   heat-­‐inactivated   fetal   calf   serum   (GIBCO/BRL,   Carlsbad,   CA,   USA),   2   mM   of   L-­‐glutamine,   100   μg/ml   streptomycin   sulphate   (Sigma)   and   100   U/ml   penicillin   (Sigma),   at   37   °C   in   a   humidified   atmosphere   of   air  containing  5  %  CO2.        

87  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

2.3.

Preparation  of  LN  incorporating  edelfosine  

LN   were   prepared   by   a   warm   oil-­‐in-­‐water   (o/w)   microemulsion   followed   by   high  shear  homogenization  and  ultrasonication.  The  lipid  phase  consisted  of  either   Compritol®  888  ATO  or  Precirol®  ATO  5  along  with  edelfosine,  while  the  aqueous   phase   consisted   of   a   2   %   Tween®   80   aqueous   solution.   The   aqueous   phase   was   heated  at  about  5  °C  above  the  melting  point  of  the  lipid  and  added  to  the  melted   lipid   phase   at   the   same   temperature.   The   mixture   was   dispersed   with   the   help   of   a   MicrosonTM   ultrasonic   cell   disruptor   (NY,   USA)   for   1   minute.   The   preformed   emulsion   was   then   homogenised   in   an   Ultraturrax®   (IKA-­‐Werke,   Germany)   for   1   minute   and   sonicated   again   with   the   MicrosonTM   ultrasonic   cell   disruptor   (NY,   USA)   for   1   minute.   The   nanoparticle   suspension   was   cooled   in   an   ice   bath   and   washed   twice   with   filtered   water   by   diafiltration   with   Amicon   Ultra-­‐15   filters   of   10,000   dalton   molecular   weight   cut-­‐off   membrane   (Millipore®,   Cork,   Ireland)   to   remove  the  excess  of  surfactant.  Nanoparticles  were  then  resuspended  in  PBS  for   animal  administration  or  in  10  %  trehalose  solution  for  freeze-­‐drying.     2.4.

Characterization  of  edelfosine  loaded  LN   2.4.1. Particle  size  and  zeta  potential  

The  average  particle  size  and  polydispersity  index  of  edelfosine  loaded  LN  were   determined   by   photon   correlation   spectroscopy   (PCS)   using   a   Zetasizer   Nano   (Malvern  Instruments,  UK).  Each  sample  was  diluted  30  fold  in  distilled  water  until   the   appropriate   concentration   of   particles   was   achieved   to   avoid   multiscattering   events.   The   obtained   homogenous   suspension   was   examined   to   determine   the   volume,   mean   diameter,   size   distribution   and   polydispersity   and   repeated   three   times  for  each  sample.  Similarly,  the  zeta  potential  was  measured  using  the  same   equipment   with   a   combination   of   Laser   Doppler   (LD)   velocimetry.   Samples   were   diluted   with   distilled   water   and   each   experiment   was   performed   in   triplicate.   All   data  are  expressed  as  a  mean  value  ±  standard  deviation.      

88  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

2.4.2. Loading  capacity   Edelfosine  was  quantified  by  an  ultra  high-­‐performance  liquid  chromatography   tandem   mass   spectrometry   (UHPLC-­‐MS/MS)   method   that   had   been   previously   validated   [19].   The   drug   was   extracted   from   a   sample   of   10   mg   of   lyophilized   nanoparticles,  to  which  1  ml  of  chloroform  was  added  in  order  to  dissolve  them.  10   μL  of  the  internal  standard  PAF  (0.2  mg/mL)  were  then  spiked  to  the  samples.  3   mL   of   methanol   were   added   to   the   mixture,   and   after   vortex   mixing   for   1   min   at   room  temperature  and  centrifuging  at  20,000  ×  g  for  10  min,  2  μL  aliquots  of  the   supernatant  were  injected  into  the  chromatographic  system.     2.5.

Hemolysis  experiments  

Erythrocytes   from   fresh   human   blood   were   separated   from   plasma   by   centrifugation   (4,000   ×   g,   7   min)   and   washed   three   times   with   PBS.   4   mL   of   the   washed   erythrocyte   suspension   were   diluted   to   100   mL   with   PBS.   1.5   mL   of   this   suspension   were   treated   with   0.5   mL   of   tested   samples:   free   edelfosine   (10   μg/mL),  edelfosine  loaded  Compritol®  and  Precirol®  nanoparticles  (10  μg/mL)  and   drug  free  Compritol®  and  Precirol®  nanoparticles.  Absorbance  was  measured  in  an   Agilent  8453  UV-­‐visible  spectrophotometer  (Agilent,  Palo  Alto,  CA,  USA)  at  540  nm   1  h  after  the  treatment.     2.6.

Animal  experiments    

Animal   handling   was   conducted   in   compliance   with   the   regulations   of   the   Ethical   Committee   of   the   University   of   Navarra   as   well   as   with   the   European   Community   Council   Directive   Ref.   86/609/EEC.   For   pharmacokinetic   studies,   BALB/c  mice  (20  g)  were  obtained  from  Harlan  Interfauna  Ibérica  S.L.  (Barcelona,   Spain).   For   efficacy   studies,   SCID   mice   (Janvier,   Genest   St   Isle,   France)   were   employed.   Animals   received   a   standard   diet   and   water   ad   libitum,   except   for   the   animals   that   received   the   oral   doses,   which   were   fasted   for   24   hours   prior   to   administration.     89  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

2.6.1. Pharmacokinetic   and   biodistribution   studies   after   intravenous   administration     An   i.v.   single   dose   of   edelfosine-­‐loaded   LN   (50   mg/kg)   was   administered   to   BALB/c   (n=8   per   group)   mice   via   the   tail   vein.   Group   1   received   Compritol®   888   ATO   LN   and   group   2,   Precirol®   ATO   5   LN.   At   various   time   points   after   administration  (0,  1,  2,  5,  8,  24  and  31  h  for  Compritol®  group  and  0,  1,  2,  5,  8,  24,   48,  72,  96,  120,  144  and  168  h  for  Precirol®  group),  blood  was  collected  in  EDTA   surface-­‐coated   tubes   and   then   centrifuged   at   2,000   ×   g   for   10   min   (4   °C)   to   separate   the   plasma   (100   μL).   Then,   animals   were   sacrificed   and   spleen,   liver,   lungs,   kidneys,   heart,   stomach   and   intestine   were   collected   and   weighed.   Tissues   were   homogenised   in   1   mL   of   PBS   pH=7.4   using   a   Mini-­‐bead   Beater   (BioSpect   Products,   Inc.,   Bartelsville,   Oklahoma,   USA)   and   centrifuged   at   10,000   ×   g   for   10   min.  Both  plasma  and  tissue  supernatants  were  collected  and  stored  at  -­‐80  °C  until   UHPLC-­‐MS/MS  analysis  was  performed.     2.6.2. Pharmacokinetic  and  biodistribution  studies  after  oral  administration   Two   BALB/c   mice   groups   were   treated   with   a   single   oral   administration   of   edelfosine-­‐loaded   LN   (edelfosine   concentration   of   50   mg/kg,   n=8   per   group).   Group   1   received   Compritol®   888   ATO   LN   and   group   2,   Precirol®   ATO   5   LN.   At   various  time  points  after  the  administration  (0,  1,  2,  5,  8,  24,  48,  72,  96,  120,  144   and   168   h   for   Compritol®   group   and   0,   1,   2,   5,   8,   24,   48,   72,   96,   120,   144,   168,   192   and  216  h  for  Precirol®  group),  blood  was  collected  in  EDTA  surface-­‐coated  tubes   and  then  centrifuged  at  2,000  ×  g  for  10  min  (4  °C)  to  collect  plasma  (100  μL).  After   sacrifice  by  cervical  dislocation,  tissues  were  collected,  weighed  and  processed  as   explained  above.       2.6.3. Lymphatic  absorption  studies   A   group   of   BALB/c   mice   received   an   oral   dose   of   both   types   of   LN   (edelfosine   concentration  of  50  mg/kg,  n=8).  24  hours  later,  an  oily  emulsion  (milk)  was  orally   administered  1  hour  prior  to  sacrifice,  in  order  to  make  the  lymph  ducts  and  nodes  

90  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

more  visible,  animals  were  sacrificed  and  mesenteric  lymph  nodes  were  extracted   and  analyzed  by  UHPLC-­‐MS/MS.     2.6.4. Efficacy  studies     Eight-­‐week   SCID   mice   were   subcutaneously   inoculated   into   the   lower   dorsum   with   1   x   107   JVM-­‐2   cells   in   100   μL   of   PBS   and   100   μL   of   Matrigel   basement   membrane   matrix   (Becton   Dickinson,   San   Jose,   CA).   Animals   received   a   standard   diet   and   water   ad   libitum.   When   tumors   were   palpable,   mice   were   randomly   assigned  to  the  treatment  groups.  Six  groups  of  mice  (n=8  per  group)  were  treated   orally:   group   1:   PBS;   group   2:   edelfosine   solution   (30   mg/kg   dissolved   in   PBS);   group   3:   edelfosine-­‐loaded   Compritol®   888   ATO   nanoparticles   (edelfosine   concentration   of   30   mg/kg);   group   4:   edelfosine-­‐loaded   Precirol®   ATO   5   nanoparticles  (edelfosine  concentration  of  30  mg/kg);  group  5:  blank  Compritol®   888   ATO   nanoparticles   (10   mg/mL   lipid   concentration);   and   group   6:   blank   Precirol®  ATO  5  nanoparticles  (10  mg/mL  lipid  concentration,  equivalent  to  a  30   mg/kg   edelfosine   dose).   The   treatments   were   administered   by   oral   gavage   every   four  days.  The  experiment  ended  when  control  group  tumors  reached  a  volume  of   5.0±0.5  cm3.  At  this  point,  animals  were  sacrificed  and  tumors  were  collected  for   the   determination   of   their   volume   and   weight.   Axilary,   inguinal   and   mesenteric   lymph  nodes  were  also  extracted  from  MCL-­‐bearing  mice  treated  with  either  free   or  vectorized  edelfosine  and  macroscopically  analyzed.     2.7.

Data  analysis    

Pharmacokinetic   analysis   was   performed   with   plasma   samples   obtained   from   experiments  with  all  mice.  All  these  plasma  concentration  data  were  analyzed  by   non-­‐compartmental   and   compartmental   analysis   using   WinNonlin   Professional   Edition   Version   2.1   (Pharsight,   Mountain   View,   CA,   USA).   The   area   under   the   plasma   concentration   vs.   time   curve   (AUC)   was   determined   using   the   log-­‐linear   trapezoidal  rule  with  extrapolation  to  infinitum  and  normalized  against  the  dose.   The   CL   value   is   the   volume   of   plasma   completely   cleared   of   a   specific   compound  

91  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

per  unit  time  by  the  organism;  it  was  calculated  by  dividing  the  dose  by  AUC.  The   maximum   plasma   concentration   (Cmax)   was   calculated   from   the   plasma   concentration-­‐time  curve  and  normalized  against  the  dose.  Oral  bioavailability  (F)   was   determined   by   ratio   of   the   dose-­‐normalized   AUC   following   oral   and   i.v.   administration.   Vss   is   the   volume   of   fluid   that   would   be   required   to   contain   the   amount   of   drug   in   the   body   if   it   were   uniformly   distributed   at   a   concentration   equal   to   that   in   the   plasma.   The   t1/2   value   refers   to   the   time   taken   for   plasma   concentration  to  fall  by  50  %,  and  it  was  determined  using  the  following  formula:   t1/2=ln  (2)·Vss/CL.     2.8.

Statistical  analysis    

The   presence   of   differences   in   tissue/plasma   ratios   and   pharmacokinetic   parameters  was  measured  by  the  Mann  Whitney  test  for  double  comparisons  using   Social   Package   of   Statistical   Sciences   (SPSS).   Student’s   t   test   was   used   for   measuring   differences   in   efficacy   and   tumor   dissemination   inhibition   studies.   A   value   of   p   <   0.05   was   considered   to   be   statistically   significant   for   all   statistical   tests.    

3. Results  and  discussion    

Traditionally,   nanoparticulated   drug   delivery   systems   have   been   formulated   using   organic   solvents   as   solubilizing   agents.   However,   toxicity   of   organic   solvents   is   a   special   concern   as   it   may   lead   to   the   dismissal   of   any   proposed   formulation   regardless   of   potential   benefits.   After   administration,   nanoparticles   are   generally   uptaken  by  macrophages  or  the  mononuclear  phagocytes  in  the  organism,  leading   to   generation   of   inflammatory   and   tissue   response.   Thus,   the   solvents   used,   degradation  products  of  the  nanoparticles,  adhesion  of  these  degradation  products   to  the  membrane  and  consequent  stimulation  of  cells  and  release  of  inflammatory   mediators   may   be   the   cause   of   the   toxicity   of   nanoparticles.   The   avoidance   of   organic   solvents   during   the   preparation   of   LN   by   the   warm   oil-­‐in-­‐water   (o/w)  

92  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

microemulsion   followed   by   high   shear   homogenization   and   ultrasonication   method   decreases   the   mentioned   potential   toxicity.   Edelfosine-­‐loaded   LN   were   produced   by   a   warm   oil-­‐in-­‐water   (o/w)   microemulsion   followed   by   high   shear   homogenization   and   ultrasonication   method,   and   freeze-­‐dried.   This   solvent-­‐free   method   does   not   entail   the   utilization   of   toxic   organic   solvents   with   obvious   implications  from  a  biocompatibility  point  of  view.    

3.1.

Particle  size,  size  distribution  and  zeta  potential  

The   peroral   route   is   the   most   preferred   route   of   administration,   but   this   path   is   limited   for   many   substances   as   they   present   poor   oral   bioavailability   due   to   biopharmaceutical   (low   solubility,   low   permeability,   and/or   instability   in   gastrointestinal   environment)   and   pharmacokinetic   (extensive   first   pass   metabolism   and/or   rapid   clearance)   drawbacks   in   their   delivery   approach.   Therefore,   the   development   of   delivery   systems   that   would   be   able   to   overcome   these   drawbacks   is   essential   to   ensure   the   effectiveness   of   such   molecules.   The   physical-­‐chemical   characteristics   of   the   developed   nanoparticles   are   compiled   in   Table  I.  All  data  are  expressed  as  mean  value  ±  standard  deviation.      

Table   I.   Average   size,   PDI,   zeta   (ζ)   potential,   encapsulation   efficiency   (EE)   and   drug   loading   of   edelfosine-­‐loaded   LN   (n=20)   prepared   by   the   warm   microemulsion   formation   followed   by   high   shear  homogenization  and  ultrasonication  method.  

ζ  Potential   (mV)  

%  EE  

Drug  loading  (μg   edelfosine/mg  form.)  

Drug-­‐free   130.6  ±  3.1   0.275  ±  0.021   Compritol®  LN  

-­‐28.6  ±  2.1  

−  

−  

Drug-­‐loaded   110.4  ±  2.1   0.241  ±  0.050   Compritol®  LN  

-­‐21.2  ±  1.5   84.68  ±  7.18  

LN  

Size  (nm)  

PDI  

Drug-­‐free   Precirol®  LN  

117.7  ±  2.4   0.243  ±  0.026  

-­‐29.1  ±  1.7  

Drug-­‐loaded   Precirol®  LN  

103.1  ±  2.9   0.231  ±  0.012  

-­‐22.4  ±  2.0   82.62  ±  5.73  

−  

17.57  ±  1.97   −   13.95  ±  0.79  

 

93  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

Drug-­‐free  LN  presented  diameters  of  130.6  ±  3.1  and  117.7  ±  2.4  for  Compritol®   and   Precirol®   LN,   respectively.   The   average   diameter   of   edelfosine-­‐loaded   Compritol®   LN   was   110.4   ±   2.1   nm,   while   drug-­‐loaded   Precirol®   LN   presented   a   mean  diameter  of  103.1  ±  2.9  nm,  suggesting  that  edelfosine  might  be  responsible   for   the   reduction   in   size   of   LN,   as   it   is   a   surfactant   structured   molecule.   These   particles   present   a   smaller   size   than   those   prepared   by   the   emulsion   formation   and  solvent  evaporation  method  previously  developed  [5].  This  method  therefore   provides  smaller  particles,  which  are  organic  solvent-­‐free,  in  less  formulation  time.   PDI   was   below   0.3   in   all   cases   indicating   that   the   LN   were   homogenous   in   size.   These   particles   present   an   appropriate   size   for   their   oral   administration,   since   sizes  below  300  nm  are  suitable  for  intestinal  transport  to  the  thoracic  duct  [20].     Zeta   (ζ)   potential   can   make   a   prediction   about   the   stability   of   colloid   dispersions.   A   high   ζ   potential   (>|30|   mV)   can   provide   an   electric   repulsion   to   avoid  the  aggregation  of  particles  [21].  In  our  case,  the  ζ-­‐potential  values  measured   in  double-­‐distilled  water  were  negative.  The  mean  ζ  potentials  of  edelfosine-­‐loaded   and  drug-­‐free  Compritol®  LN  were  -­‐21.2  ±  1.5  mV  and  -­‐28.6  ±  2.1  mV,  respectively.   Precirol®   LN   showed   similar   values   of   -­‐22.4   ±   2.0   mV   and   -­‐29.1   ±   1.7   mV   for   edelfosine-­‐loaded   and   drug-­‐free   Precirol®   LN,   respectively.   The   incorporation   of   edelfosine  into  LN  had  little  influence  on  the  zeta  potentials  of  nanoparticles.  This   negative  surface  charge  could  be  due  to  the  presence  of  oleic  acid  traces  in  Tween®   80   on   the   particle   surface,   forming   a   denser   surfactant   film,   and   thus   eliciting   increased   electrophoretic   mobility.   Besides,   the   steric   impediment   of   Tween®   80   might  be  another  effect  which  would  increase  the  stability  of  colloidal  dispersions   [22].    

3.2.

Loading  capacity  

It  is  well  known  that  the  crystalline  state  in  the  LN  structure  leads  to  faster  drug   expulsion.   However,   lattice   defects   of   the   lipid   structure   offer   space   to   accommodate   the   drugs   [23].   As   a   result,   the   structure   of   less   ordered   arrangement  in  the  nanoparticles  should  be  beneficial  to  the  drug  loading  capacity,   as  in  the  case  of  the  particles  developed  in  this  work.    

94  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

The   entrapment   efficiency   of   edelfosine   in   the   LN   prepared   by   the   warm   microemulsion   followed   by   high   shear   homogenization   and   ultrasonication   was   similar   for   nanoparticles   prepared   with   both   types   of   lipids   (Table   I).   Nanoparticles   formulated   using   Compritol®   encapsulated   84.68   ±   7.18   %   of   edelfosine   (equivalent   to   17.57   ±   1.97   μg   edelfosine/mg   formulation),   while   Precirol®   LN   encapsulated   82.62   ±   5.73   %   of   the   drug   (13.95   ±   0.79   μg   edelfosine/mg  formulation).  This  high  encapsulation  efficiency  is  likely  to  be  due   to  the  partially  amorphous  state  of  the  lipids  in  the  formulation,  which  allows  more   edelfosine  to  be  incorporated  among  lipid  chains  [5].     3.3.

Pharmacokinetic  

characterization  

and  

biodistribution  

after  

intravenous  administration     Figures   1A   and   1B   show   the   concentration   of   edelfosine   in   mouse   plasma   plotted   against   time   after   a   single   i.v.   administration   of   edelfosine-­‐loaded   LN   (concentration  ranging  30  -­‐  60  mg/kg)  to  BALB/c  mice.    

  Figure  1.  Time-­‐concentration  curve  data  of  edelfosine  after  a  single  intravenous  administration  of   edelfosine  loaded  (A)  Compritol®  and  (B)  Precirol®  LN  to  BALB/c  mice  (n=8,  mean  ±  S.D.)    

Dose-­‐normalized   pharmacokinetic   analysis   of   edelfosine   in   blood   plasma   showed   a   Cmax   of   approximately   0.3   μg/mL   for   both   types   of   LN,   showing   no   statistical   differences   between   them.   All   obtained   pharmacokinetic   parameters   were  dose-­‐normalized  and  are  listed  in  Table  II.      

95  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

Table   II.   Comparison   of   pharmacokinetic   parameters   of   edelfosine   after   intravenous   administration   of   edelfosine   loaded   LN   (50   mg/kg   bw,   n=8   per   group,   mean  ±  SD).  No  statistical  differences  were  found  among  parameters  p>0.05.   Parameters  

Compritol®  888  ATO  LN  i.v.   Precirol®  ATO  5  LN  i.v.   administration   administration  

t½α  (h)  

0.395  ±  0.124  

0.505  ±  0.151  

t½β  (h)  

16.970  ±  5.775  

23.718  ±  17.743  

Cmax  /  D  (μg/mL/μg)  

0.290  ±  0.087  

0.341  ±  0.044  

CL  (L/h/kg)  

0.105  ±  0.021  

0.065  ±  0.023  

Vss  (L/kg)  

1.668  ±  0.730  

1.313  ±  0.838  

MRT  (h)  

17.154  ±  6.517  

26.096  ±  20.598  

AUCinf  /  D  (μg/mL/μg)  

0.573  ±  0.053  

0.894  ±  0.399  

                   

Plasma  concentration-­‐time  data  of  edelfosine  in  LN  were  well  described  by  bi-­‐ exponential   functions   following   i.v.   administration   of   both   types   of   LN.   The   distribution  half-­‐lives  (t1/2α)  of  the  two  formulations  were  around  0.4  h,  while  the   elimination   half-­‐lives   (t1/2β)   were   17   h   for   Compritol®   LN   and   23   h   for   Precirol®   LN,   suggesting   a   much   slower   elimination   of   these   last   nanoparticles.   These   parameters  are  higher  than  those  of  the  liposomes  described  by  Bhamra  et  al.  [4],   indicating  that  these  LN  circulate  in  plasma  for  a  longer  period  of  time.   The   rest   of   the   pharmacokinetic   parameters   showed   no   statistical   differences   between  the  two  types  of  LN.  The  mean  systemic  CL  and  Vss  values  for  edelfosine-­‐ loaded   LN   were   around   0.08   L/h/kg   and   1.5   L/kg,   respectively.   When   edelfosine   was   loaded   into   liposomes,   the   Vss   was   7   times   lower   than   this   value,   suggesting   that   LN   allow   a   broader   distribution   of   the   drug   in   the   body.   There   was   little   variability   in   most   of   the   values   of   the   pharmacokinetic   parameters,   indicating   a   well-­‐controlled   and   reproducible   study,   except   for   the   elimination   phase   half-­‐life   value.  AUC  values  were  between  0.6  and  0.9  μg·h/mL/μg,  similar  to  those  obtained   in  a  previous  research  work  [2]  after  the  i.v.  administration  of  edelfosine  solution,  

96  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

indicating  that  the  i.v.  administration  of  edelfosine  in  LN  presents  a  bioavailability   of  100  %.     Figure   2   depicts   the   scattering   of   the   drug   through   the   mouse   body   after   i.v.   administration  of  edelfosine-­‐loaded  LN,  expressed  as  tissue/plasma  ratios.                             Figure   2.   Tissue/plasma   concentration   ratios   of   edelfosine   after   a   single   intravenous   dose   of   edelfosine-­‐loaded  Compritol®  and  Precirol®  LN  to  BALB/c  mice  (n=8)  

  Whichever  the  type  of  nanoparticle  employed,  the  highest  accumulations  of  the   drug  were  achieved  in  kidney,  intestine  and  liver,  followed  by  spleen,  stomach  and   lung,  with  no  statistical  differences  between  the  ratios.       3.4.

Pharmacokinetic   characterization   and   biodistribution   after   oral  

administration   Pharmacokinetic   studies   were   performed   after   a   single   oral   administration   of   50  mg/kg  of  edelfosine-­‐loaded  in  LN.  This  oral  dose  was  well  tolerated  by  the  mice   and  no  hemolytic  side  effects  or  body  weight  loss  was  observed  (data  not  shown).   Figure   3   shows   the   concentration   of   edelfosine   in   mouse   plasma   plotted   against   97  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

time  after  a  single  oral  administration  of  edelfosine-­‐loaded  LN  to  BALB/c  mice.  The   endpoint   of   the   experiment   was   the   day   after   the   concentration   of   edelfosine   in   plasma  reached  0.5  μg/mL.  

 

 

 

 

  Figure   3.   Time-­‐plasma   concentration   curve   data   of   edelfosine   obtained   with   the   WinNonLin   program  after  a  single  oral  administration  of  edelfosine  solution  and  edelfosine  loaded  Compritol®   and  Precirol®  LN  to  mice  (n=8  per  group)    

It  can  be  observed  that  unlike  edelfosine  solution,  drug-­‐loaded  LN  enhanced  the   absorption  of  the  drug  maintaining  detectable  concentrations  for  over  seven  days.   As  a  result,  the  maintenance  times  in  plasma  achieved  with  the  developed  LN  were   much  longer  than  those  observed  by  different  studies  recently  published  with  the   same  Compritol®  lipid,  which  did  not  last  longer  than  24  h  [24,  25].     All   dose-­‐normalized   pharmacokinetic   parameters   obtained   after   the   administration  of  edelfosine  in  LN  are  summarized  in  Table  III.          

98  

Chapter  3.  Lipid  nanoparticles  loaded  with  alkyl-­‐lysophospholipid  edelfosine:   Pharmacokinetic  profile,  biodistribution  studies  and  in  vivo  efficacy  against  MCL    

  Table   III.   Comparison   of   pharmacokinetic   parameters   of   edelfosine   after   oral   administration   of   edelfosine   loaded   LN   (50   mg/kg   bw,   n=8   per   group,   mean   ±   SD).   Asterisks   indicate   significantly   different  values  between  the  two  types  of  LN,  p99.9%) was provided by

Praxair (Madrid, Spain). Chemical structures of edelfosine and PAF are shown in Fig. 1. 2.2. Instruments and analysis conditions 2.2.1. HPLC–MS method This method is based on a previous method developed in our group [14]. The apparatus used for the HPLC analysis was a Model 1100 series LC coupled with an atmospheric pressure–electrospray ionization (ESI) single quadrupole mass spectrometer equipped with a collision-induced dissociation cell (HP 1100 with MSD VL, Waldbronn, Germany). Separation was carried out at 50 ◦ C on a reversed-phase, 150 mm × 3 mm column packed with C18 , 5 !m silica reversed-phase particles (Gemini® ) obtained from Phenomenex® (Torrance, CA, USA). This column was preceded by a reversed-phase, C18 , 5 !m guard column (SecurityGuardTM , 20 mm × 4 mm, Phenomenex® , Torrance, CA, USA). The mobile phase was a mixture of methanol–1% formic acid (95:5, v/v). Separation was achieved by isocratic solvent elution at a flow rate of 0.5 mL/min. The MS was operated in the positive ESI mode. The detection of edelfosine and the internal standard was performed by selected ionization monitoring (SIM) mode. ESI-MS conditions were as follows: source temperature 350 ◦ C, capillary voltage 4 kV, and collision-induced dissociation voltage 140 V. Nitrogen was used as the desolvation gas with a flow rate of 12 L/min and a pressure of 30 psi (1 psi = 6894.76 Pa). Optimization of the interface variables, such as gas flows and voltages was done manually during direct infusion of 10 !g/mL of the target analyte dissolved in methanol. The spectrometer was programmed to monitor both the ion of edelfosine at m/z 524.4 and platelet activating factor at m/z 574.4. Under these conditions, edelfosine and I.S. were eluted at 3.65 and 3.50 min, respectively. Data acquisition and analysis were performed with a Hewlett-Packard computer using the ChemStation G2171 AA program (Agilent, Palo Alto, CA, USA). 2.2.2. UHPLC–MS/MS method The UHPLC system was composed of an Acquity UPLCTM system (Waters Corp., Milford, MA, USA) with thermostatized autosampler and column compartment. Separation was carried out on an Acquity UPLCTM BEH C18 column (50 mm × 2.1 mm, 1.7 !m; Waters Corp., Milford, MA, USA) with isocratic elution using a mobile phase composed of 5% of a 1% formic acid aqueous solution and 95% of methanol. Column temperature was maintained at 50 ◦ C. The flow rate was set at 0.5 mL/min. The autosampler was conditioned at 4 ◦ C and the injection volume was 2 !L using partial loop mode for sample injection. Triple-quadrupole tandem mass spectrometric detection was performed on an AcquityTM TQD mass spectrometer (Waters Corp., Milford, MA, USA) with an electrospray ionization (ESI) interface. The mass spectrometer operated in positive mode was set up for multiple reaction monitoring (MRM) to monitor the transition of m/z 524.3 → 104.2 for edelfosine and the transition of m/z 552.3 → 184.2 for the internal standard, with the dwell time of 0.1 s per transition. To optimize the MS parameters, standard solutions of both the analyte and internal standard were infused into the mass spectrometer. The following optimized MS parameters were employed: 4 kV capillary voltage, 60 V cone voltage for edelfosine and 30 V for the internal standard, 150 ◦ C source temperature and 350 ◦ C desolvation temperature. Nitrogen was used for the desolvation and as cone gas at a flow rate of 650 and 50 L/h, respectively. Argon was used as the collision gas. The optimized collision energy for edelfosine was 30 and 20 eV for the internal standard. Under these conditions, edelfosine and I.S. were eluted at 1.23 ± 0.01 and 1.19 ± 0.02 min, respectively. Data acquisition and analysis were performed using the MassLynxTM

    260  

Annex  II.  Comparative  study  of  a  HPLC–MS  assay  versus  an  UHPLC–MS/MS  for  anti-­‐tumoral   alkyl  lysophospholipid  edelfosine  determination  in  both  biological  samples  and  in  lipid   nanoparticulate  systems  

A. Estella-Hermoso de Mendoza et al. / J. Chromatogr. B 877 (2009) 4035–4041

4037

Fig. 1. Chemical structure of (A) Edelfosine and (B) platelet activation factor.

NT 4.1 software with QuanLynxTM program (Waters Corp., Milford, MA, USA). 2.3. Preparation of standard and quality control (QC) samples 2.3.1. Standard solutions and QC samples for plasma Stock solutions of edelfosine were prepared in methanol. The stock solution of PAF was prepared in methanol at a concentration of 0.2 mg/mL. For the HPLC, two calibration ranges had to be established for sample quantitation between 0.1 and 75 !g/mL. The calibration curves for concentration range from 0.1 to 1 !g/mL were

prepared by adding 50 !L of the standard solutions of 0.2, 1 and 2 !g/mL to 100 !L of blank mouse plasma. Effective concentrations of edelfosine were 0.1, 0.5 and 1 !g/mL. Calibration curves for a concentration range from 1 to 75 !g/mL were prepared by adding 50 !L of the standard solutions of 2, 30, 60 and 150 !g/mL to mouse plasma. Effective concentrations of edelfosine were 1, 15, 30 and 75 !g/mL. The QC samples were pooled at concentrations of 0.1, 5, 10 and 50 !g/mL. For the UHPLC, calibration curves for a concentration range from 0.0075 to 75 !g/mL were prepared by adding 50 !L of the standard solutions of 0.015, 6, 60 and 150 !g/mL to 100 !L of blank mouse plasma. Effective concentrations of edelfosine were

Fig. 2. Representative chromatograms of: (A) blank mouse plasma sample; (B) edelfosine resulting from the analysis of mouse plasma obtained at 1 min after intravenous administration of 200 !g of edelfosine; (C) Platelet Activating Factor (PAF) resulting from the analysis of mouse plasma after intravenous administration of 200 !g of edelfosine (PAF concentration = 0.2 mg/mL); (D) blank mouse plasma sample; (E) plasma sample after an intravenous administration of 200 !g of edelfosine to a BALB/c mouse and (F) Platelet Activating Factor (PAF) resulting from the analysis of mouse plasma after intravenous administration of 200 !g of edelfosine (PAF concentration = 0.2 mg/mL).

    261  

Annex  II.  Comparative  study  of  a  HPLC–MS  assay  versus  an  UHPLC–MS/MS  for  anti-­‐tumoral   alkyl  lysophospholipid  edelfosine  determination  in  both  biological  samples  and  in  lipid   nanoparticulate  systems  

4038

A. Estella-Hermoso de Mendoza et al. / J. Chromatogr. B 877 (2009) 4035–4041

0.0075, 3, 30 and 75 !g/mL. The QC samples were pooled at concentrations of 0.075, 0.75, 15 and 60 !g/mL. The spiked plasma samples (standard and QC) were then processed following the application of the extraction procedure.

at 20,000 × g for 10 min, 200 !L of the supernatant were mixed with 800 !L of methanol and then, 5 and 2 !L aliquots were injected into the HPLC and UHPLC systems, respectively. 2.5. Method validation

2.3.2. Standard solutions and QC samples for tissues Stock solutions of edelfosine were prepared in methanol. The stock solution of PAF was prepared in methanol at a concentration of 0.2 mg/mL. For the HPLC, calibration curves were prepared by adding 50 !L of the standard solutions of 0.4, 4, 8, 16 and 63.5 !g/mL to tissue homogenate in buffer (see Section 2.6). Effective concentrations of edelfosine were 0.2, 2, 4, 8 and 31.75 !g/mL. The QC samples were pooled at concentrations of 0.1, 1.72, 17.19 and 31.75 !g/mL. For the UHPLC, calibration curves for a concentration range from 0.0075 to 75 !g/mL were prepared by adding 50 !L of the standard solutions of 0.015, 6, 60 and 150 !g/mL to the tissue homogenate. Effective concentrations of edelfosine were 0.0075, 3, 30 and 75 !g/mL. The QC samples were pooled at concentrations of 0.075, 0.75, 15 and 60 !g/mL. The spiked tissue samples (standard and QC) were then processed after application of extraction procedure. 2.3.3. Standard solutions and QC samples for lipid nanoparticles The stock solution of PAF was prepared in methanol at a concentration of 0.2 mg/mL. Stock solutions of edelfosine were prepared in methanol. For the HPLC, effective concentrations of edelfosine were 1, 15, 30 and 75 !g/mL. The QC samples were pooled at concentrations of 1, 5, 10 and 50 !g/mL. For the UHPLC, effective concentrations of edelfosine were 0.0075, 3, 30 and 75 !g/mL. The QC samples were pooled at concentrations of 0.075, 0.75, 15 and 60 !g/mL. The edelfosine standards and QCs were then injected into the chromatographic system. 2.4. Sample preparation 2.4.1. Plasma samples Blood was collected in EDTA surface-coated tubes and then centrifuged at 2000 × g for 10 min (4 ◦ C) to separate the plasma. A portion of 100 !L of mouse plasma was transferred to a 1.5-mL tube and then 10 !L of PAF (0.2 mg/mL), used as internal standard (I.S.) were spiked to the samples. Then, 190 !L of mobile phase (methanol–1% formic acid (95:5, v/v)) were added and the mixture was vortex-mixed at room temperature for 1 min for precipitation. After centrifuging at 20,000 × g for 10 min, 200 !L of the supernatant were mixed with 800 !L of methanol and then, 5 and 2 !L aliquots were injected into the HPLC and UHPLC systems, respectively. 2.4.2. Tissue samples A portion of 100 !L of tissue homogenate in buffer (see Section 2.6) was transferred to a 1.5-mL tube and then, 10 !L of I.S. (0.2 mg/mL) were spiked to the samples and 10 !L of 10% TCA were added to the mixture and vortex-mixed for 10 s for protein precipitation. Finally, 180 !L of mobile phase were added to the mixture. After vortex mixing for 1 min at room temperature and centrifuging at 20,000 × g for 10 min, 200 !L of the supernatant were mixed with 800 !L of methanol and then, 5 !L and 2 !L aliquots were injected into the HPLC and UHPLC systems, respectively. 2.4.3. Lipid nanoparticles A sample of 10 mg of lyophilized nanoparticles was weighed in a 10-mL tube and then, 1 mL of chloroform was added in order to dissolve the nanoparticles. 10 !L of I.S. (0.2 mg/mL) were spiked to the samples. Then, 3 mL of mobile phase were added to the mixture. After vortex mixing for 1 min at room temperature and centrifuging

The selectivity of the assay was determined by the individual analysis of blank samples. The retention times of endogenous compounds in the matrix were compared with those of edelfosine and PAF. LOD was defined as the sample concentration resulting in a peak area of three times the noise level. LOQ was defined as the lowest drug concentration, which can be determined with an accuracy and precision < 20%. In this work LOD of the assay method was determined by analysis of the peak baseline noise in ten blank samples. Plasma samples were quantified using the internal standard method. Standard curves were calculated using linear least squares regression between theoretical edelfosine concentration on calibrator samples and the chromatographic peak area ratios of edelfosine to that of the internal standard. To evaluate linearity, calibrator samples were prepared and analyzed in duplicate on 3 separate days. Accuracy and precision were also determined by replicate measurements (n = 6) of quality control samples at four concentration levels on five different validation days. The accuracy was expressed as (real concentration − theoretical concentration)/(theoretical concentration) × 100 and the precision by the CV (%) of the measured concentration values obtained after analysis of the quality control samples with different nominal concentration values. The absolute extraction recoveries of edelfosine at three QC levels were evaluated by measuring the samples as described above and comparing the peak areas of the edelfosine and the I.S., and then comparing with those obtained from direct injection of the compounds dissolved in the supernatant of the processed blank biological samples (plasma or the different tissues). The matrix effect was evaluated by comparing the peak area of the analyte dissolved in the reconstituted residues of processed blank plasma with the standard solutions at the same concentration dissolved in mobile phase. The matrix effect was evaluated at three different concentration levels, with three samples analyzed in each set. The matrix effect of the internal standard was evaluated at the concentration in plasma samples using the same method. 2.6. Application of the method The present method has been successfully applied to the quantitation of edelfosine in biological matrices and lipid nanocarriers. To demonstrate the reliability of this method for the study of edelfosine pharmacokinetics, this assay was applied to the quantitation of edelfosine in plasma samples obtained from 6 BALB/c mice treated with an intravenous dose of 200 !g of an edelfosine solution. Tissue distribution of edelfosine was also studied. Blood samples were withdrawn at 0, 1, 2, 5, 8 and 24 h postadministration in EDTA surface-coated tubes and then centrifuged at 2000 × g for 10 min (4 ◦ C) to separate the plasma (100 !L). Plasma was stored frozen (−80 ◦ C) until analysis. Then, the animals were sacrificed and the spleen, liver, lungs, kidneys, heart, brain, stomach and intestine were collected and weighed. Tissue samples were homogenized in 1 mL of Phosphate Buffered Saline (PBS) using a Mini-bead Beater (BioSpect Products, Inc., Bartelsville, OK, USA) and centrifuged at 10,000 × g for 10 min. Supernatant was separated and stored frozen (−80 ◦ C) until analysis. This method was also employed for the assessment of encapsulation efficiency in lipid nanoparticles. Compritol® 888 ATO is the lipid employed for the formulation of nanoparticles. The method for

    262  

Annex  II.  Comparative  study  of  a  HPLC–MS  assay  versus  an  UHPLC–MS/MS  for  anti-­‐tumoral   alkyl  lysophospholipid  edelfosine  determination  in  both  biological  samples  and  in  lipid   nanoparticulate  systems  

4039

A. Estella-Hermoso de Mendoza et al. / J. Chromatogr. B 877 (2009) 4035–4041 Table 1 Standard calibration curves of edelfosine in plasma, tissue and particulate homogenate samples calculated by the HPLC method. Range (!g/mL) 0.1–1 Plasma 1–75 Kidney Liver Lung Heart Spleen Brain Stomach Intestine Particulate homogenate

0.2–31.75 0.2–31.75 0.2–31.75 0.2–31.75 0.2–31.75 0.2–31.75 0.2–31.75 0.2–31.75 1–75

Regression equation

r

y = 0.1342x + 0.0001 y = 0.1013x + 0.0051 y = 0.1159x + 0.0018 y = 0.0221x − 0.0471 y = 0.0345x + 0.0972 y = 0.0267x − 0.0474

0.999 1.000 0.999 0.999 0.999 0.999

y = 0.1387x − 0.0149 y = 0.1327x − 0.0059 y = 0.1410x − 0.0603 y = 0.1862x − 0.0038 y = 0.1721x − 0.0662 y = 0.1519x − 0.0715 y = 0.1251x + 0.0069 y = 0.1244x + 0.0008 y = 0.1276x + 0.0096

0.999 0.999 0.999 1.000 1.000 0.999 0.999 1.000 0.999

r: correlation coefficient.

lipid nanoparticle formulation is the emulsion formation/solvent evaporation method. Briefly, lipid and edelfosine were dissolved in chloroform and homogenized by ultrasonication with an aqueous solution of 2% Tween® 80. The obtained emulsion was then subjected to mechanical stirring for the organic solvent evaporation and consequent lipid nanoparticle formation. Particles were then centrifuged at 4500 × g for 10 min using an Amicon Ultra-15 filter device and washed twice with distilled water. The obtained particular suspension was fast frozen at −80 ◦ C for at least 3 h and freeze-dried in order to store it at 4 ◦ C [24]. 3. Results and discussion A sensitive method for edelfosine detection in plasma and tissue samples was needed for their concentration-time course measurements during dose escalation and other pharmacokinetic preclinical studies. To achieve this aim it is critical to optimize the chromatographic conditions to obtain symmetrical peak shapes and a short chromatographic analysis time with high sensitivity and selectivity. Previously, we developed a HPLC–MS method for the quality control of edelfosine drug delivery systems and the edelfosine quantitation in cell internalization studies. Under these chromatographic conditions edelfosine is eluted as tailing and band-broadening chromatographic peaks (values of USP asymmetry factor between 1.1 and 1.3), with insufficient chromatographic efficiency to measure this drug at low concentrations [14]. Chemically, edelfosine is an anionic amphiphilic compound that is positively charged at acidic pH, which is the pH of the mobile phase. The observed peak tailing may be a result of an ionic interaction of residual package silanols and positively charged nitrogen of edelfosine. One of the first efforts was the attempt to reduce the value of the limit of quantitation while maintaining the employed extraction procedure. In our previous work, edelfosine determination was performed with polymerically bounded C18 reversed-phase narrow-bore column packed with double encapped spherical silica particles (Alltima® ). To overcome the technical difficulties observed, a substantial improvement of the chromatographic performance of the method is the only valid solution. For the HPLC method, we have used a Gemini® column instead of the Alltima® package. Gemini® column has been developed with a technology that grafts additional silica–organic layers onto the surface of the internal base silica. These additional layers protect the particle from ionic interactions with ionized compounds, such as edelfosine at acidic pH, and as a result, sharp, symmetrically chromatographic peaks are obtained.

Table 2 Standard calibration curves of edelfosine in plasma, tissue and particulate homogenate samples calculated by the UHPLC method. Range (!g/mL)

Regression equation

R

Plasma

0.0075–75

y = 0.03831x − 0.0004 y = 0.04029x + 0.0005 y = 0.03551x − 0.0007

0.999 0.999 0.999

Kidney Liver Lung Heart Spleen Brain Stomach Intestine Particulate homogenate

0.0075–75 0.0075–75 0.0075–75 0.0075–75 0.0075–75 0.0075–75 0.0075–75 0.0075–75 0.0075–75

y = 0.0321x + 0.0004 y = 0.0413x + 0.0002 y = 0.0429x − 0.0005 y = 0.0526x + 0.0001 y = 0.0520x − 0.0003 y = 0.0397x + 0.00007 y = 0.0762x + 0.0003 y = 0.0447x + 0.0005 y = 0.0313x + 0.0009

0.999 0.999 0.999 0.999 0.999 1.000 0.999 0.999 0.999

r: correlation coefficient.

On the other hand, and according to the van Deemter equation, one helpful way to improve the efficiency and analysis time of the HPLC column is to decrease the particle size. However, the improved column efficiency gained from using small particles comes along with a tremendous increase in the column pressure, which is prohibitive for traditional HPLC hardware, but not for UHPLC hardware, which can easily resist pressure values up to 15,000 psi [15,19]. For an ideal comparison, we would prefer to use columns with identical chemistry. However, at the time of the study, columns packed with Gemini® C18 particles < 2 !m were unavailable. Thus, Acquity BEH UPLCTM C18 columns packed with bridged ethylsiloxane-silica particles were employed. Fig. 2 shows the typical chromatograms of (A) a blank and (B) a spiked plasma sample with edelfosine and (C) the internal standard analyzed by the HPLC technique and (D) a blank and (E) a spiked plasma sample with edelfosine and (F) the internal standard analyzed by the UHPLC technique. The typical retention time for edelfosine and internal standard was 3.65 and 3.50 min for the HPLC–MS method, and 1.23 and 1.19 min for edelfosine and internal standard, respectively, for the UHPLC–MS/MS method. Compared with HPLC, UHPLC reduced the retention times threefold on average. The resolution between the chromatographic peaks of the edelfosine and internal standard was 0.08 and 0.09 for the HPLC–MS and the UPLC–MS/MS methods, respectively. Therefore, we can affirm that there is little difference in column selectivity between the two types of package employed. The HPLC asymmetry factor for edelfosine was 2, whereas it was 1.2 for the UHPLC method. The main improvement in the chromatographic behavior has been reflected in method sensibility. In fact, a sixfold increase in the LOQ value was observed for the UHPLC–MS/MS method compared to that of the HPLC–MS method, which made edelfosine quantitation possible in small samples, such as that obtained in pharmacokinetic studies in mice, after intravenous administration of sub-milligram doses. Validation data for edelfosine quantitation by the HPLC and UHPLC methods are compared in Tables 1–3. Assay performance of the present methods was assessed by all the following criteria: selectivity, linearity, accuracy, precision, LOD, LOQ, applicability to quantitation of edelfosine in different matrices and quantitation of edelfosine in lipid nanoparticles. Selectivity was assessed by the comparison of the chromatograms of six different batches of blank mouse plasma with the corresponding spiked plasma. There was no relevant interference from endogenous substances observed at the retention times of the analytes, as can be seen in Fig. 2. The HPLC–MS assay exhibited linearity divided into two intervals between the response (y) and the corresponding concentration of edelfosine (x) from 0.1 to 1 !g/mL in the small concentration interval and from 1 to 75 !g/mL in the high concentration

    263  

Annex  II.  Comparative  study  of  a  HPLC–MS  assay  versus  an  UHPLC–MS/MS  for  anti-­‐tumoral   alkyl  lysophospholipid  edelfosine  determination  in  both  biological  samples  and  in  lipid   nanoparticulate  systems  

 

4040

A. Estella-Hermoso de Mendoza et al. / J. Chromatogr. B 877 (2009) 4035–4041

Table 3 Accuracy, precision and between- and within-day measured concentrations for analysis of edelfosine by the HPLC and UHPLC methods. Conc. (!g/mL)

Accuracy (bias %)

Precision (%RSD) Between-day

Measured conc. (!g/mL, mean ± SD) Within-day

Between-day

Within-day

HPLC method QC1 QC2 QC3 QC4

0.1 5 10 50

4.88 7.13 −5.83 −0.62

7.77 7.11 5.57 2.04

5.42 4.23 1.66 3.22

0.10 5.22 9.50 50.16

± ± ± ±

0.01 0.37 0.53 1.02

0.09 5.49 9.28 48.90

± ± ± ±

0.01 0.23 0.15 1.58

UHPLC method QC1 QC2 QC3 QC4

0.075 0.75 15 60

−6.84 −4.06 6.49 2.73

10.91 7.20 7.70 4.59

12.23 8.33 6.60 3.72

0.069 0.70 15.80 62.06

± ± ± ±

0.007 0.05 1.19 2.84

0.070 0.73 15.97 61.29

± ± ± ±

0.008 0.06 1.03 2.28

interval for plasma samples. Tissue and nanoparticulate system samples presented linearity from 0.2 to 31.75 !g/mL and from 1 to 75 !g/mL, respectively. The UHPLC–MS/MS method showed a much higher sensitivity and calibration range than the obtained with the HPLC method. A linear range was achieved for all types of samples from 0.0075 to 75 !g/mL and the limit of quantitation was 0.0075 !g/mL. Results are shown in Tables 1 and 2. For each point of calibration standards, the concentrations were back-calculated from the equation of the regression curves and relative standard deviations (%RSD) were measured. %RSD did not exceed 15% in any case. For all calibration curves, linear regression provided r values greater than 0.999. As can be seen in Tables 1 and 2, the slope of the calibration curves from each tissue sample was similar to that obtained after the chromatographic analysis of plasma calibrator samples in a similar concentration range. It is clear that the developed method is adequate to quantify edelfosine in different biological samples, where an adequate extraction procedure has been applied. The lower limit of quantitation for edelfosine with the HPLC method was 0.1 !g/mL (S/N ≥ 5) with 5 !L injected into the chromatographic column with accuracy within ±20% and %RSD lower than 20%. The UHPLC method showed a limit of quantitation of 0.0075 !g/mL with 2 !L injected into the chromatographic column, presenting similar accuracy values. Compared with the HPLC method, the present UHPLC method gave sixfold higher sensitivity. The high sensitivity of the UHPLC method could be attributed to the peak sharpness produced by the column package and the lower analyte dilution in the column. To determine recovery, concentrations of edelfosine and PAF in extracted plasma and tissue QC samples were compared to standards prepared in blank matrix extract. The recovery was eval-

Fig. 3. Time-concentration curve data of edelfosine over 24 h after single dose intravenous administration of 200 !g (10 mg/kg) to BALB/c mice (n = 6, Mean ± S.D.) calculated by the HPLC method.

uated in triplicate and presented acceptable values ranging from 85.11% and 100.93%, and from 83.64% to 107.48% for edelfosine and internal standard, respectively, by the HPLC method, and from 95.25% to 99.35% and from 93.43% to 93.79% for edelfosine and PAF, respectively, by the UHPLC method. Matrix ionization suppression is considered to be a problem when using the protein precipitation method for sample preparation. Nevertheless, this method has been chosen as the sample preparation procedure in our work due to its simplicity and the lack of impact on the accuracy of the assay. The matrix effect values of edelfosine and the I.S. in both plasma and tissue homogenate samples were 15.55 ± 1.79% and 9.03 ± 3.96% for the HPLC method, and 6.17 ± 2.77% and 3.71% for the UHPLC method. It is interesting to note that the matrix effect is slightly reduced when the UHPLC analysis is performed. This may be due to the fact that the signal suppression is diminished drastically in the UHPLC–MS/MS method compared with the HPLC–MS method. Similar data were obtained by the HPLC and UHPLC methods after accuracy and precision evaluation (Table 3). Accuracy values were within acceptable limits ranging between −6.84% and 7.13%. The results for intra-batch and inter-batch precision for the samples ranged between 1.66% and 12.23%. The precision and accuracy of the present method is in accordance with the criteria for the analysis of biological samples according to the guidance of the FDA, where the precision (expressed as %RSD) determined at each concentration level is required not to exceed 15%. The applicability of this method has been demonstrated in vivo by the determination of edelfosine in plasma samples from BALB/c mice treated with 200 !g of edelfosine. Fig. 3 depicts the concentration of edelfosine in mouse plasma plotted against time after a single-dose intravenous administration of 200 !g of edelfosine (10 mg/kg) to BALB/c mice determined by the HPLC method. Edelfosine in blood plasma showed a Cmax of 50.7 ± 28.1 !g/mL and a Cmin of 2.5 ± 1.3 !g/mL, 24 h after intravenous administration. Tissue levels of edelfosine were also measured and compared to those

Fig. 4. Tissue distribution of edelfosine 24 h after a single intravenous administration of 200 !g to mice (10 mg/kg) calculated by the UHPLC method.

  264  

Annex  II.  Comparative  study  of  a  HPLC–MS  assay  versus  an  UHPLC–MS/MS  for  anti-­‐tumoral   alkyl  lysophospholipid  edelfosine  determination  in  both  biological  samples  and  in  lipid   nanoparticulate  systems  

A. Estella-Hermoso de Mendoza et al. / J. Chromatogr. B 877 (2009) 4035–4041

of plasma. Fig. 4 depicts the distribution of edelfosine to different organs in mice after intravenous administration of 200 !g of edelfosine determined by the UHPLC method. No drug was detectable in the heart and very little was found in the brain. The edelfosine levels present in the kidney and intestine were statistically very significant compared to the plasma levels of edelfosine (p < 0.01). Edelfosine levels in the lung and heart were statistically different from plasma levels (p < 0.05). No significant differences were found between plasma and liver, spleen, brain and stomach levels (p > 0.05). The drug was also extracted from previously formulated lipid nanoparticles and quantified. Nanoparticulate systems made of lipid material Compritol® 888 ATO showed an encapsulation efficiency of about 95%. The same samples were analyzed with both the HPLC–MS and UHPLC–MS/MS method to ensure the interchangeability of both methods. The edelfosine results obtained from liver samples analyzed by the UHPLC–MS/MS method were highly correlated (r2 = 0.91; y = 0.9706x + 0.2123) with those from the HPLC–MS method. A good relationship between both techniques was found over the concentration range of 0.2–31.75 !g/mL for the HPLC–MS and UHPLC–MS/MS. 4. Conclusion Two liquid chromatographic methods, an HPLC–MS method and a UHPLC–MS/MS method for the bio-analysis of edelfosine were developed and evaluated. The UHPLC–MS/MS method developed in this work was more sensitive for the quantitation of edelfosine in plasma, tissue and lipid nanoparticles than the HPLC–MS method. Under the UPLC conditions we are able to achieve a shorter chromatographic run time while still avoiding a matrix ion suppression problem. UHPLC and HPLC are valuable methods for the determination of the pharmacokinetic behavior of edelfosine and bio-distribution in mice after intravenous administration of a dose of 10 mg/kg of edelfosine and the quality control of lipid nanoparticulate systems. Acknowledgements The authors would like to thank Caja Navarra Foundation and the Spanish Ministry of Science and Innovation (SAF2007-61261,

4041

PCT-090100-2007-27, SAF2008-02251 and RD06/0020/1037) for their support. Ander Estella-Hermoso de Mendoza is supported by the research grant from the Department of Education of the Basque Government (BFI06.37). References [1] C. Gajate, F. Mollinedo, Curr. Drug. Metab. 3 (2002) 491. [2] F. Mollinedo, C. Gajate, S. Martin-Santamaria, F. Gago, Curr. Med. Chem. 11 (2004) 3163. [3] W.J. Houlihan, M. Lohmeyer, P. Workman, S.H. Cheon, Med. Res. Rev. 15 (1995) 157. [4] P.G. Munder, O. Westphal, Chem. Immunol. 49 (1990) 206. [5] F. Mollinedo, J.L. Fernandez-Luna, C. Gajate, B. Martin-Martin, A. Benito, R. Martinez-Dalmau, M. Modolell, Cancer Res. 57 (1997) 1320. [6] G.A. Ruiter, M. Verheij, S.F. Zerp, W.J. van Blitterswijk, Int. J. Radiat. Oncol. Biol. Phys. 49 (2001) 415. [7] C. Gajate, F. Mollinedo, Blood 109 (2007) 711. [8] W.R. Vogler, W.E. Berdel, R.B. Geller, J.A. Brochstein, R.A. Beveridge, W.S. Dalton, K.B. Miller, H.M. Lazarus, Adv. Exp. Med. Biol. 416 (1996) 389. [9] R. Bhamra, L.E. Bolcsak, I. Ahmad, J. Schupsky, P. Roberts, R. Stevens, C. Cavanaugh, C.E. Swenson, Anticancer Drugs 14 (2003) 481. [10] J. Kotting, N.W. Marschner, W. Neumuller, C. Unger, H. Eibl, Prog. Exp. Tumor Res. 34 (1992) 131. [11] J. Coene, M. Ghis, P. Van den Eeckhout, P. Van den Bossche, J. Sandra, J. Chromatogr. 553 (1991) 285. [12] J. Coene, E. Van den Eeckhout, P. Herdewijn, P. Sandra, J. Chromatogr. 612 (1993) 21. [13] W.F. Steelant, E.A. Bruyneel, M.M. Mareel, E.G. Van den Eeckhout, Anal. Biochem. 227 (1995) 246. [14] M.J. Blanco-Prieto, M.A. Campanero, F. Mollinedo, J. Chromatogr. B: Analyt. Technol. Biomed. Life Sci. 810 (2004) 85. [15] M.I. Churchwell, N.C. Twaddle, L.R. Meeker, D.R. Doerge, J. Chromatogr. B: Analyt. Technol. Biomed. Life Sci. 825 (2005) 134. [16] I.S. Lurie, J. Chromatogr. A 1100 (2005) 168. [17] I. Citova, L. Havlikova, L. Urbanek, D. Solichova, L. Novakova, P. Solich, Anal. Bioanal. Chem. 388 (2007) 675. [18] D. Guillarme, D.T. Nguyen, S. Rudaz, J.L. Veuthey, Eur. J. Pharm. Biopharm. 66 (2007) 475. [19] Y. Hsieh, C.J. Duncan, S. Lee, M. Liu, J. Pharm. Biomed. Anal. 44 (2007) 492. [20] J. Olsovska, M. Jelinkova, P. Man, M. Koberska, J. Janata, M. Flieger, J. Chromatogr. A 1139 (2007) 214. [21] N. Stephanson, A. Helander, O. Beck, J. Mass Spectrom. 42 (2007) 940. [22] J.C. Van De Steene, W.E. Lambert, J. Am. Soc. Mass Spectrom. 19 (2008) 713. [23] K. Yu, D. Little, R. Plumb, B. Smith, Rapid Commun. Mass Spectrom. 20 (2006) 544. [24] A. Estella-Hermoso de Mendoza, M. Rayo, F. Mollinedo, M.J. Blanco-Prieto, Eur. J. Pharm. Biopharm. 68 (2008) 207.

  .   265  

 

         

 

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

 

Annex  III   In  vitro  and  in  vivo  selective  antitumor  activity  of   edelfosine  against  mantle  cell  lymphoma  and  chronic   lymphocytic  leukemia  involving  lipid  rafts   Faustino  Mollinedo1,  Janis  de  la  Iglesia-­‐Vicente1,  Consuelo  Gajate1,2,  Ander  Estella-­‐ Hermoso  de  Mendoza3,  Janny  A.  Villa-­‐Pulgarin1,  Mercè  de  Frias4,  Gaël  Roué5,  Joan   Gil4,  Dolors  Colomer5,  Miguel  A.  Campanero6,  and  Maria  J.  Blanco-­‐Prieto3    

1Instituto  de  Biología  Molecular  y  Celular  del  Cáncer,  Centro  de  Investigación  del  

Cáncer,  CSIC-­‐Universidad  de  Salamanca,  Campus  Miguel  de  Unamuno,  E-­‐37007   Salamanca,  Spain;  2Unidad  de  Investigación,  Hospital  Universitario  de  Salamanca,   Campus  Miguel  de  Unamuno,  E-­‐37007  Salamanca,  Spain;  3Departamento  de   Farmacia  y  Tecnología  Farmacéutica,  Facultad  de  Farmacia,  Universidad  de   Navarra,  E-­‐31008  Pamplona,  Spain;  4Departament  de  Ciències  Fisiològiques  II,   Institut  d'Investigació  Biomèdica  de  Bellvitge  (IDIBELL)–Universitat  de  Barcelona,  E-­‐ 08907  L'Hospitalet  de  Llobregat,  Spain  ;  5Unitat  d’Hematopatologia,  Hospital  Clínic,   Institut  d’Investigacions  Biomèdiques  August  Pi  i  Sunyer  (IDIBAPS)-­‐Universitat  de   Barcelona,  E-­‐08036  Barcelona,  Spain;  6Servicio  de  Farmacología  Clínica,  Clínica   Universitaria,  E-­‐31080  Pamplona,  Spain;.    

         

Clinical  Cancer  Research,  2010.  16(7):  p.  2046-­‐2054

267  

 

   

 

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

Clinical Cancer Research

Cancer Therapy: Preclinical

In vitro and In vivo Selective Antitumor Activity of Edelfosine against Mantle Cell Lymphoma and Chronic Lymphocytic Leukemia Involving Lipid Rafts Faustino Mollinedo1, Janis de la Iglesia-Vicente1, Consuelo Gajate1,2, Ander Estella-Hermoso de Mendoza3, Janny A. Villa-Pulgarin1, Mercè de Frias5, Gaël Roué6, Joan Gil5, Dolors Colomer6, Miguel A. Campanero4, and Maria J. Blanco-Prieto3

Abstract Purpose: Mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL) remain B-cell malignancies with limited therapeutic options. The present study investigates the in vitro and in vivo effect of the phospholipid ether edelfosine (1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine) in MCL and CLL. Experimental Design: Several cell lines, patient-derived tumor cells, and xenografts in severe combined immunodeficient mice were used to examine the anti-MCL and anti-CLL activity of edelfosine. Furthermore, we analyzed the mechanism of action and drug biodistribution of edelfosine in MCL and CLL tumor-bearing severe combined immunodeficient mice. Results: Here, we have found that the phospholipid ether edelfosine was the most potent alkyllysophospholipid analogue in killing MCL and CLL cells, including patient-derived primary cells, while sparing normal resting lymphocytes. Alkyl-lysophospholipid analogues ranked edelfosine > perifosine ≫ erucylphosphocholine ≥ miltefosine in their capacity to elicit apoptosis in MCL and CLL cells. Edelfosine induced coclustering of Fas/CD95 death receptor and rafts in MCL and CLL cells. Edelfosine was taken up by malignant cells, whereas normal resting lymphocytes hardly incorporated the drug. Raft disruption by cholesterol depletion inhibited drug uptake, Fas/CD95 clustering, and edelfosine-induced apoptosis. Edelfosine oral administration showed a potent in vivo anticancer activity in MCL and CLL xenograft mouse models, and the drug accumulated dramatically and preferentially in the tumor. Conclusions: Our data indicate that edelfosine accumulates and kills MCL and CLL cells in a rather selective way, and set coclustering of Fas/CD95 and lipid rafts as a new framework in MCL and CLL therapy. Our data support a selective antitumor action of edelfosine. Clin Cancer Res; 16(7); 2046–54. ©2010 AACR.

Authors' Affiliations: 1Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Cientificas–Universidad de Salamanca, Campus Miguel de Unamuno; 2Unidad de Investigación, Hospital Universitario de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain; 3Departamento de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Navarra; 4Servicio de Farmacología Clínica, Clínica Universitaria, Pamplona, Spain; 5 Departament de Ciències Fisiològiques II, Institut d'Investigació Biomèdica de Bellvitge–Universitat de Barcelona, L'Hospitalet de Llobregat, Spain; and 6Unitat d'Hematopatologia, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer–Universitat de Barcelona, Barcelona, Spain Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Faustino Mollinedo, Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Cientificas–Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain. Phone: 34-923-294806; Fax: 34-923-294795; E-mail: [email protected] or Consuelo Gajate, Unidad de Investigación, Hospital Universitario de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain. E-mail: [email protected]. doi: 10.1158/1078-0432.CCR-09-2456 ©2010 American Association for Cancer Research.

2046

Chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) are two major B-cell–derived neoplasias for which current therapy is not satisfactory, leading in most cases to relapse and eventually to a fatal outcome. This lack of efficient therapy underscores the need for a continued search for novel chemotherapeutic agents. CLL is the most common adult leukemia and is characterized by the progressive accumulation of mature CD5+ B lymphocytes in the peripheral blood, bone marrow, and secondary lymphoid organs. New treatment combinations have incorporated the use of purine analogue (fludarabine)–based regimens together with monoclonal antibodies rituximab (anti-CD20) and alemtuzumab (anti-CD52), leading to improved complete response rates and prolonged progression-free survival, but a long-term survival benefit has not been shown (1, 2). MCL is characterized by the chromosomal translocation t(11;14)(q13;q32), resulting in the overexpression of cyclin D1 in mature B

Clin Cancer Res; 16(7) April 1, 2010

  269  

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

Antitumor Activity of Edelfosine against MCL and CLL

Translational Relevance Mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL) lack effective therapy. Synthetic alkyl-lysophospholipid analogues constitute a family of promising anticancer drugs, including miltefosine, perifosine, edelfosine, and erucylphosphocholine, which promote apoptosis in a variety of tumor cells. Here, we have found that edelfosine behaves as the most potent alkyl-lysophospholipid analogue in inducing cell death in MCL and CLL cells through coclustering of Fas/CD95 and rafts. Edelfosine induced a higher apoptotic response than perifosine in MCL and CLL patient-derived cells. Oral administration of edelfosine showed a strong in vivo anti-MCL and anti-CLL activity in xenograft mouse models. The drug accumulated in a dramatic and preferential way in the tumor, leading to drastic tumor regression. Our data reported here show a rather selective action of edelfosine against tumor cells and provide the proof of principle and rationale for further clinical evaluation of edelfosine to improve patient outcome in MCL and CLL.

lymphocytes that have a striking tendency to disseminate throughout the body (3). MCL is an aggressive lymphoma with a poor survival outcome and a median survival time of 3.5 years. Conventional chemotherapeutic regimens have been the standard treatment of MCL until the recent incorporation of rituximab, which increases overall survival as well as the response rate and duration. The introduction of stem cell transplantation improves survival, although this therapeutic modality is only applied to younger and fit patients (4). Currently, allogeneic bone marrow transplantation represents the only therapy with the potential for a curative approach, although it is associated with a high rate of complications (4). Therefore, development of novel therapeutic strategies is urgently needed to improve survival in patients with the above B-cell malignancies. In the last years, new strategies have been developed that target crucial cellular pathways, and proteasome inhibition with bortezomib has recently been approved in relapsed/ refractory MCL (5). MCL and CLL cells share a relatively low proliferative index and a poor apoptotic rate (6, 7), and therefore, the transforming event is likely a failure in death regulation rather than a loss of growth control. This implies that a therapeutic potential for these diseases may lie in potentiating apoptosis. Synthetic alkyl-lysophospholipid (ALP) analogues constitute a family of promising anticancer drugs, including edelfosine, miltefosine, perifosine, and erucylphosphocholine, which act at the cell membrane level (8–12). The phospholipid ether edelfosine (1-O-octadecyl-2-Omethyl-rac-glycero-3-phosphocholine, ET-18-OCH3), considered the ALP prototype, induces selectively apoptosis in tumor cells (13) by recruiting Fas/CD95 death receptor in

www.aacrjournals.org

membrane raft clusters (13–18). Edelfosine was the first antitumor agent shown to act through lipid rafts (15), and these membrane domains have been recently involved in the chemotherapeutic action of some additional antitumor agents (17, 19–23). Lipid rafts are membrane microdomains highly enriched in cholesterol and sphingolipids, and the proteins located in these microdomains are limited in their ability to freely diffuse over the plasma membrane, affecting protein function (24). Recent findings suggest that lipid rafts act as scaffolds, where Fas/ CD95 and downstream signaling molecules are recruited to trigger apoptosis (16–18, 21, 25). On these grounds, we hypothesized that MCL and CLL, showing a defective cell death control, could be considered stark examples for the particular proapoptotic features of ALPs. Thus, we addressed the possibility that these B-cell malignancies could be treated by these agents, and examined the putative involvement of lipid rafts in their antitumor action against MCL and CLL.

Materials and Methods Drugs. Edelfosine was from Inkeysa and Apointech. Miltefosine was from Calbiochem. Perifosine and erucylphosphocholine were from Zentaris. Cell lines and primary cells. Detailed information on the cell culture conditions for human MCL (JVM-2 and Z-138) and CLL (EHEB) cell lines and primary cells from CLL and MCL patients is included in Supplementary Data. Apoptosis assay. Quantitation of apoptotic cells was determined by flow cytometry as the percentage of cells in the sub-G1 region (hypodiploidy) in cell cycle analysis as previously described (26). To analyze apoptosis in CLL/MCL patient-derived samples, 5 × 105 cells were incubated for 48 h with the indicated agents. Cells were then washed in Annexin-binding buffer and incubated in 50 μL Annexin-binding buffer with allophycocyanin-conjugated anti-CD3 and phycoerythrinconjugated anti-CD19 antibodies from Becton Dickinson for 10 min in the dark. Cells were then diluted with Annexin-binding buffer to a volume of 150 μL and incubated with 1 μL FITC-labeled Annexin V (Bender MedSystems) for 15 min in the dark. A total of 10,000 stained cells were then analyzed by flow cytometry on a FACSCalibur flow cytometer using CellQuest software (Becton Dickinson). Western blot. Proteins (50 μg) were separated on 12% SDS-PAGE and transferred onto Immobilon-P membranes (Millipore). Membranes were probed with anti– cyclin D1 (DCS-6, Cell Signaling Technology) and anti–β-actin (Sigma) antibodies. Antibody binding was detected using the enhanced chemiluminescence detection system (Amersham). Confocal microscopy. Cells were settled onto poly- Llysine–coated slides and analyzed with a Zeiss LSM 510 laser scan confocal microscope for membrane raft and Fas/CD95 visualization using FITC-labeled cholera toxin B subunit (Sigma) and anti-human Fas/CD95 SM1/1 IgG2a mouse monoclonal antibody (Bender MedSystems)

Clin Cancer Res; 16(7) April 1, 2010

2047

  270  

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

Mollinedo et al.

Fig. 1. Induction of apoptosis in MCL and CLL cells by ALPs. MCL (JVM-2 and Z-138) and CLL (EHEB) cell lines were incubated for the indicated times with 10 μmol/L of the distinct ALPs edelfosine, perifosine, miltefosine, and erucylphosphocholine (Erucyl-PC; A) or for 24 h with different concentrations of the ALPs (B). Apoptosis was then quantitated as percentage of cells in the sub-G1 region by flow cytometry. Untreated control cells were run in parallel. Data are means ± SE of four independent determinations. C, cells were untreated or treated with 10 μmol/L perifosine or edelfosine for the indicated times and analyzed by Western blot with anti–cyclin D1 and anti–β-actin antibodies. Immunoblotting of β-actin was used as an internal control for equal protein loading in each lane. Blots are representative of three experiments done.

followed by CY3-conjugated anti-mouse antibody (Pharmacia), as described (15). Colocalization assays were analyzed by excitation of both fluorochromes in the same section. Negative controls, lacking the primary antibody or using an irrelevant antibody, showed no staining. Edelfosine uptake. Drug uptake was measured as described previously (13) after incubating 106 cells with 10 nmol [3H]edelfosine for 2 h in RPMI 1640/10% fetal bovine serum and subsequent exhaustive washing (six times) with PBS + 2% bovine serum albumin to eliminate the loosely cell surface–bound ether lipid. [3H]Edelfosine (specific activity, 42 Ci/mmol) was synthesized by

2048

Clin Cancer Res; 16(7) April 1, 2010

tritiation of the 9-octadecenyl derivative (Amersham Buchler). Cholesterol depletion. Cells (2.5 × 105/mL) were pretreated with 2.5 mg/mL methyl-β-cyclodextrin (MCD) for 30 min at 37°C in serum-free medium. Cells were then washed thrice and resuspended in complete culture medium before edelfosine addition. Xenograft mouse model. CB17–severe combined immunodeficient (SCID) mice (Charles River Laboratories), kept and handled according to institutional guidelines, complying with Spanish legislation under a 12/12-h light/dark cycle at a temperature of 22°C, received a standard diet

Clinical Cancer Research

  271  

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

Antitumor Activity of Edelfosine against MCL and CLL

and acidified water ad libitum. CB17-SCID mice were inoculated s.c. into their lower dorsum with 107 Z-138 or EHEB cells in 100 μL PBS and 100 μL Matrigel basement membrane matrix (Becton Dickinson). When tumors were palpable, mice were randomly assigned into cohorts of 8 to 10 mice each, receiving a daily oral administration of edelfosine (30 mg/kg) or an equal volume of vehicle (PBS). The shortest and longest diameter of the tumor were measured with calipers at 2- to 5-d intervals, and tumor volume (mm3) was calculated using the following standard formula: (the shortest diameter)2 × (the longest diameter) × 0.5. Animals were sacrificed, according to institutional guidelines, when the diameter of their tumors reached 3 cm or when significant toxicity was observed. Animal body weight and any sign of morbidity were monitored. Drug treatment lasted for 21 d (MCL) and 34 d (CLL), and mice were killed 24 h after the last drug administration. Then, tumor xenografts were extirpated, measured, and weighed, and a necropsy analysis involving tumors and distinct organs was carried out. Plasma/tissue extraction procedure for edelfosine biodistribution studies. MCL- or CLL-bearing SCID mice were treated with a daily oral administration of edelfosine (30 mg/kg) for 21 d (MCL) or 34 d (CLL). Twenty-four hours after the last drug oral administration, blood was collected in EDTA surface-coated tubes and then centrifuged at 2,000 × g for 15 min (4°C) to collect plasma (100 μL). Then, animals were sacrificed, and distinct organs and tumors were collected and weighed. Tissues and tumors were homogenized in 1-mL PBS (pH 7.4) using a Mini-bead Beater (BioSpect Products, Inc.) and centrifuged at 10,000 × g for 10 min. Both plasma and tissue supernatants

Table 1. Selective killing of patient-derived CLL cells by edelfosine, sparing normal cells Treatment

% Cell viability CD19+ CLL cells CD3+ T cells

Edelfosine (10 μmol/L) Edelfosine (20 μmol/L) Perifosine (10 μmol/L) Perifosine (20 μmol/L) Staurosporine (0.5 μmol/L)

60.3 43.7 73.1 61.0 17.8

± ± ± ± ±

5.6 4.9 4.8 3.7 6.7

97.8 86.5 98.4 90.9 22.5

± ± ± ± ±

1.9 3.2 1.5 2.8 7.5

NOTE: Primary lymphocyte cultures from CLL patients were incubated with edelfosine or perifosine for 48 h at the indicated concentrations. Staurosporine was used as a positive control of apoptosis. Percentage of cell viability was measured as nonapoptotic CD3+/CD19− T cells or CD3−/CD19+ B cells from CLL samples in Annexin V analysis by flow cytometry. Untreated control CD19+ and CD3+ cells were run in parallel and showed a cell viability of >93% and >98%, respectively. Data are shown as mean values ± SE of five independent CLL patients.

were collected and stored at −80°C until high-performance liquid chromatography–mass spectrometry (HPLC-MS) analysis was done. Ten micrograms of platelet-activating factor (1 mg/mL), used as internal standard, were added onto 100 μL of plasma or tissue/tumor supernatant. A mixture (190 μL) of 1% formic acid/methanol was added to precipitate proteins. Samples were vortexed for 1 min, and after centrifugation (20,000 × g, 10 min), 25 μL of the supernatant were analyzed by HPLC-MS. Quantitative determination of edelfosine by HPLC-MS analysis. The technique used was a slight modification of a previously described method (27) and is described in detail in Supplementary Data. Statistical analysis. All values are expressed as means ± SE. Between-group statistical differences were assessed using the Mann-Whitney test or the Student's t test. A P value of 85% in tumor weight and volume in both MCL and CLL animal models (Fig. 5B). Organ examination at necropsy did not reveal any apparent toxicity (data not shown), and there was an evident difference between the highly vascularized tumors from drug-free mice and the pale poorly vascularized tumors from edelfosine-treated mice (Fig. 5C). In addition, MCL tumors were bulky in drug-free mice but resulted rather flat after edelfosine treatment (Fig. 5C). No significant differences in mean body weight were observed between drug-treated and control animals during the in vivo assay (3-5% of body weight loss in the treated groups versus control groups). A drug biodistribution study showed that edelfosine dramatically accumulated in the MCL and CLL tumors (Fig. 5D). Tumor/plasma concentration ratio of edelfosine in the tumor was significantly higher than that detected in both kidney and liver after completion of the experiment in MCL and CLL animal models (Fig. 5D), with a drug mean concentration in plasma of 5.64 μg/ mL. In the CLL animal model, we examined the content of edelfosine in a wide variety of distinct organs and found that the drug was dramatically accumulated in the tumor compared with lung, heart, spleen, liver, intestine, or kidney (Fig. 5D). Taken together, our data indicate a preferential accumulation of edelfosine in the tumor.

Discussion The data reported here show that edelfosine behaves as the most potent ALP in killing MCL and CLL cells via a raft-mediated process. Our data indicate that edelfosine is a powerful antitumor agent against MCL and CLL as assessed by in vitro, ex vivo, and in vivo evidences. In addition, we found a rather selective and dramatic accumulation of edelfosine in MCL and CLL tumor cells in animal models. Here, we found that edelfosine induces the recruitment of Fas/CD95 death receptor in raft aggregates in MCL and CLL cells. Raft disruption by cholesterol depletion in MCL and CLL cells inhibited both edelfosine uptake and druginduced apoptosis, as well as Fas/CD95 clustering, thus suggesting a major role of rafts in the uptake and antitumor action of edelfosine. Previous reports have shown that MCL and CLL cells express Fas/CD95, but a deficient apoptotic response to the external stimulation of Fas/CD95 by agonistic anti-Fas/CD95 antibodies was reported (7, 31). Unlike the natural ligand FasL/CD95L or agonistic antiFas/CD95 antibodies that act through their interaction with

Clin Cancer Res; 16(7) April 1, 2010

2051

  274  

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

Mollinedo et al.

the exogenous portion of the Fas/CD95 death receptor, edelfosine induces activation of Fas/CD95 from within the cell independently of its ligand (16, 32). We have previously found that edelfosine is even more efficient than FasL/CD95L in promoting programmed cell death through Fas/CD95 activation by its recruitment in membrane rafts enriched in downstream signaling molecules (14, 16–18, 25, 33). Thus, edelfosine might induce Fas/CD95 activation, although the receptor is not triggered by its natural ligand or agonistic antibodies. Using distinct MCL and CLL xenograft mouse models, we found that edelfosine accumulates in high amounts in the tumor tissue and shows a remarkable antitumor activity, leading to dramatic tumor regression. In addition, we consistently found in the MCL and CLL xenograft

animal models that tumors became smaller and poorly vascularized. This could be in agreement with reports showing an antiangiogenic effect of edelfosine (34, 35). Thus, further insight into the effect of edelfosine on angiogenesis and how this action affects cancer development is warranted. Following edelfosine oral administration in non–tumorbearing SCID mice, we have recently found a rather wide drug distribution pattern to several tissues, including lung, spleen, intestine, liver, and kidney (36). In this study, we also found that edelfosine showed a preferential accumulation in the tumor in a MCL-bearing mouse animal model (36). Now, we have largely extended this initial study and analyzed the in vivo effect of edelfosine in MCL and CLL animal models. Interestingly, we found here that when

Fig. 5. Edelfosine inhibits human MCL and CLL cell growth in vivo. CB17-SCID mice were inoculated s.c. with 107 Z-138 or EHEB cells. Daily oral administration of edelfosine (30 mg/kg, n = 10 for MCL animal model and n = 8 for CLL animal model) started after the development of a palpable tumor. Tumor size was recorded every 2 to 5 d. A, edelfosine significantly (P < 0.01; from day 15 of treatment until the end of the experiment) inhibited MCL and CLL tumor growth compared with the control group treated with vehicle (PBS, n = 10 for MCL animal model and n = 8 for CLL animal model). Data are means ± SE (n = 10, MCL; n = 8, CLL). B, after completion of the in vivo assay (22 d for MCL and 35 d for CLL), control and edelfosine-treated mice were sacrificed and tumors were measured in the distinct mice. The tumor size and weight values of each single animal (dots) and the average values of each experimental group (horizontal bars) are shown. Asterisks indicate that tumor weight and size values were significantly lower after edelfosine treatment, compared with control drug-free mice, at P < 0.01 (**). C, remarkable growth inhibition of MCL and CLL tumors was observed after edelfosine treatment (30 mg/kg). D, tissue/plasma concentration ratios, in liver, kidney, and tumor, of edelfosine after daily oral administration of edelfosine (30 mg/kg) for 3 wk in MCL-bearing SCID mice (mean ± SE, n = 5), and tissue/plasma concentration ratios, in lung, heart, spleen, liver, intestine, kidney, and tumor, of edelfosine after daily oral administration of edelfosine (30 mg/kg) for 34 d in CLL-bearing SCID mice (mean ± SE, n = 5). Asterisks indicate that the tumor/plasma ratio values are significantly different from the other tissue/plasma ratios at P < 0.01 (**).

2052

Clin Cancer Res; 16(7) April 1, 2010

Clinical Cancer Research

  275  

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

Antitumor Activity of Edelfosine against MCL and CLL

SCID mice contained a MCL or CLL tumor, edelfosine distribution was dramatically and significantly shifted toward the tumor (tissue/plasma concentration ratios >16; P < 0.01), suggesting a preferential tumor location for edelfosine. Our herein reported in vivo data, together with our present and previous in vitro determinations in a wide number of malignant and normal cells (14, 16, 17), suggest a rather selective edelfosine uptake and cytotoxic action in tumor cells. The selective action of edelfosine on tumor cells supports its low toxicity. We did not find any apparent damage in the distinct organs analyzed following necropsy analysis in the in vivo studies reported here. Lack of toxicity of edelfosine in a rat model has been reported, including no significant cardiotoxicity, hepatotoxicity, or renal toxicity (37). Our biodistribution data in the murine models reported here showed a mean concentration of edelfosine in plasma of 5.64 μg/mL (10.77 μmol/L; edelfosine molecular mass, 523.7). Thus, the herein reported in vitro effects, rendered by 10 μmol/L edelfosine, were detected at a pharmacologically relevant drug concentration. Our data constitute the first in vitro and in vivo evidence for the antitumor action of edelfosine in MCL and CLL, two hematologic malignancies with poor survival outcome. Taken together, the results reported here provide the proof of principle and rationale for further clinical evaluation of edelfosine to improve patient outcome in MCL and CLL. The results reported here also highlight the involvement of lipid rafts in the action of edelfosine on B-cell malignancies, such as MCL and CLL.

Disclosure of Potential Conflicts of Interest No potential conflicts of interest were disclosed.

Acknowledgments We thank Alberto Gomez-Alonso, Javier Garcia-Criado, and Jose F. Martin-Martin for their help in the in vivo assays and for allowing us to use their animal facilities during part of this study.

Grant Support Ministerio de Ciencia e Innovación grants SAF2005-04293, SAF20068850, SAF2007-61261, SAF2007-60964, PCT-090100-2007-27, PS09/ 01915, and SAF2008-02251; Red Temática de Investigación Cooperativa en Cáncer grants RD06/0020/0014, RD06/0020/0097, and RD06/0020/ 1037 (Instituto de Salud Carlos III, cofunded by the Fondo Europeo de Desarrollo Regional of the European Union); Fondo de Investigación Sanitaria and European Commission grant FIS-FEDER 06/0813; Junta de Castilla y León (CSI01A08, GR15-Experimental Therapeutics and Translational Oncology Program, and Biomedicine Project 2009); Fundación de Investigación Médica Mutua Madrileña; Fundación “la Caixa” grant BM05-30-0; Caja Navarra Foundation; Department of Health of the Government of Navarra (“Ortiz de Landázuri, 2009” project); and AGAUR-Generalitat de Catalunya grant 2005SGR-00549. C. Gajate is supported by the Ramón y Cajal Program from the Ministerio de Ciencia e Innovación of Spain. A. Estella-Hermoso de Mendoza is supported by Department of Education of the Basque Government research grant BFI06.37. M. de Frias is a recipient of a fellowship from the AGAUR-Generalitat de Catalunya. G. Roué holds a Miguel Servet research contract from Instituto de Salud Carlos III. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Received 09/08/2009; revised 12/27/2009; accepted 12/30/2009; published OnlineFirst 03/16/2010.

References 1.

Christian BA, Lin TS. Antibody therapy for chronic lymphocytic leukemia. Semin Hematol 2008;45:95–103. 2. Montserrat E, Moreno C. Chronic lymphocytic leukaemia: a short overview. Ann Oncol 2008;19 Suppl 7:vii320–5. 3. Jares P, Colomer D, Campo E. Genetic and molecular pathogenesis of mantle cell lymphoma: perspectives for new targeted therapeutics. Nat Rev Cancer 2007;7:750–62. 4. Schmidt C, Dreyling M. Therapy of mantle cell lymphoma: current standards and future strategies. Hematol Oncol Clin North Am 2008;22:953–63. 5. Leonard JP, Williams ME, Goy A, et al. Mantle cell lymphoma: biological insights and treatment advances. Clin Lymphoma Myeloma 2009;9:267–77. 6. Rummel MJ, de Vos S, Hoelzer D, Koeffler HP, Hofmann WK. Altered apoptosis pathways in mantle cell lymphoma. Leuk Lymphoma 2004;45:49–54. 7. Proto-Siqueira R, Panepucci RA, Careta FP, et al. SAGE analysis demonstrates increased expression of TOSO contributing to Fasmediated resistance in CLL. Blood 2008;112:394–7. 8. Gajate C, Mollinedo F. Biological activities, mechanisms of action and biomedical prospect of the antitumor ether phospholipid ET18-OCH3 (edelfosine), a proapoptotic agent in tumor cells. Curr Drug Metab 2002;3:491–525. 9. Jendrossek V, Handrick R. Membrane targeted anticancer drugs: potent inducers of apoptosis and putative radiosensitisers. Curr Med Chem Anti-Canc Agents 2003;3:343–53. 10. Mollinedo F, Gajate C, Martin-Santamaria S, Gago F. ET-18-OCH3 (edelfosine): a selective antitumour lipid targeting apoptosis through intracellular activation of Fas/CD95 death receptor. Curr Med Chem 2004;11:3163–84. 11. Vink SR, van Blitterswijk WJ, Schellens JH, Verheij M. Rationale and

www.aacrjournals.org

12. 13.

14.

15.

16.

17.

18.

19.

20.

clinical application of alkylphospholipid analogues in combination with radiotherapy. Cancer Treat Rev 2007;33:191–202. Mollinedo F. Antitumor ether lipids: proapoptotic agents with multiple therapeutic indications. Expert Opin Ther Patents 2007;17:385–405. Mollinedo F, Fernandez-Luna JL, Gajate C, et al. Selective induction of apoptosis in cancer cells by the ether lipid ET-18-OCH3 (edelfosine): molecular structure requirements, cellular uptake, and protection by Bcl-2 and Bcl-XL. Cancer Res 1997;57:1320–8. Gajate C, Fonteriz RI, Cabaner C, et al. Intracellular triggering of Fas, independently of FasL, as a new mechanism of antitumor ether lipid-induced apoptosis. Int J Cancer 2000;85:674–82. Gajate C, Mollinedo F. The antitumor ether lipid ET-18-OCH3 induces apoptosis through translocation and capping of Fas/CD95 into membrane rafts in human leukemic cells. Blood 2001;98:3860–3. Gajate C, Del Canto-Janez E, Acuna AU, et al. Intracellular triggering of Fas aggregation and recruitment of apoptotic molecules into Fasenriched rafts in selective tumor cell apoptosis. J Exp Med 2004;200: 353–65. Gajate C, Mollinedo F. Edelfosine and perifosine induce selective apoptosis in multiple myeloma by recruitment of death receptors and downstream signaling molecules into lipid rafts. Blood 2007;109: 711–9. Gajate C, Gonzalez-Camacho F, Mollinedo F. Involvement of raft aggregates enriched in Fas/CD95 death-inducing signaling complex in the antileukemic action of edelfosine in Jurkat cells. PLoS One 2009;4:e5044. Lacour S, Hammann A, Grazide S, et al. Cisplatin-induced CD95 redistribution into membrane lipid rafts of HT29 human colon cancer cells. Cancer Res 2004;64:3593–8. Delmas D, Rebe C, Micheau O, et al. Redistribution of CD95, DR4 and DR5 in rafts accounts for the synergistic toxicity of resveratrol

Clin Cancer Res; 16(7) April 1, 2010

2053

  276  

Annex  III.  In  vitro  and  in  vivo  selective  antitumor  activity  of  edelfosine  against  mantle  cell   lymphoma  and  chronic  lymphocytic  leukemia  involving  lipid  rafts    

Mollinedo et al.

21.

22.

23.

24. 25.

26.

27.

28.

2054

and death receptor ligands in colon carcinoma cells. Oncogene 2004;23:8979–86. Gajate C, Mollinedo F. Cytoskeleton-mediated death receptor and ligand concentration in lipid rafts forms apoptosis-promoting clusters in cancer chemotherapy. J Biol Chem 2005;280:11641–7. Mollinedo F, Gajate C. Fas/CD95 death receptor and lipid rafts: new targets for apoptosis-directed cancer therapy. Drug Resist Updat 2006;9:51–73. Reis-Sobreiro M, Gajate C, Mollinedo F. Involvement of mitochondria and recruitment of Fas/CD95 signaling in lipid rafts in resveratrolmediated antimyeloma and antileukemia actions. Oncogene 2009; 28:3221–34. Simons K, Toomre D. Lipid rafts and signal transduction. Nat Rev Mol Cell Biol 2000;1:31–9. Gajate C, Gonzalez-Camacho F, Mollinedo F. Lipid raft connection between extrinsic and intrinsic apoptotic pathways. Biochem Biophys Res Commun 2009;380:780–4. Gajate C, Santos-Beneit AM, Macho A, et al. Involvement of mitochondria and caspase-3 in ET-18-OCH3-induced apoptosis of human leukemic cells. Int J Cancer 2000;86:208–18. Blanco-Prieto MJ, Campanero MA, Mollinedo F. Quantitative determination of the antitumor alkyl ether phospholipid edelfosine by reversed-phase liquid chromatography-electrospray mass spectrometry: application to cell uptake studies and characterization of drug delivery systems. J Chromatogr B Analyt Technol Biomed Life Sci 2004;810:85–92. Gajate C, Barasoain I, Andreu JM, Mollinedo F. Induction of apoptosis in leukemic cells by the reversible microtubule-disrupting agent 2-methoxy-5-(2′,3′,4′-trimethoxyphenyl)-2,4,6-cycloheptatrien-1one: protection by Bcl-2 and Bcl-XL and cell cycle arrest. Cancer Res 2000;60:2651–9.

Clin Cancer Res; 16(7) April 1, 2010

29. Schon A, Freire E. Thermodynamics of intersubunit interactions in cholera toxin upon binding to the oligosaccharide portion of its cell surface receptor, ganglioside GM1. Biochemistry 1989;28:5019–24. 30. Harder T, Scheiffele P, Verkade P, Simons K. Lipid domain structure of the plasma membrane revealed by patching of membrane components. J Cell Biol 1998;141:929–42. 31. Plumas J, Jacob MC, Chaperot L, Molens JP, Sotto JJ, Bensa JC. Tumor B cells from non-Hodgkin's lymphoma are resistant to CD95 (Fas/Apo-1)-mediated apoptosis. Blood 1998;91:2875–85. 32. Mollinedo F, Gajate C. FasL-independent activation of Fas. In: Wajant H, editor. Fas signaling, chapter 2. Georgetown (TX): Landes Bioscience and Springer Science; 2006, p. 13–27. 33. Mollinedo F. Death receptors in multiple myeloma and therapeutic opportunities. In: Lonial S, editor. Myeloma therapy. Pursuing the plasma cell, chapter 25. Totowa (NJ): Humana Press; 2008, p. 393–419. 34. Candal FJ, Bosse DC, Vogler WR, Ades EW. Inhibition of induced angiogenesis in a human microvascular endothelial cell line by ET18-OCH3. Cancer Chemother Pharmacol 1994;34:175–8. 35. Zerp SF, Vink SR, Ruiter GA, et al. Alkylphospholipids inhibit capillary-like endothelial tube formation in vitro: antiangiogenic properties of a new class of antitumor agents. Anticancer Drugs 2008;19:65–75. 36. de Mendoza AE, Campanero MA, de la Iglesia-Vicente J, Gajate C, Mollinedo F, Blanco-Prieto MJ. Antitumor alkyl ether lipid edelfosine: tissue distribution and pharmacokinetic behavior in healthy and tumor-bearing immunosuppressed mice. Clin Cancer Res 2009;15: 858–64. 37. Mollinedo F, Gajate C, Morales AI, et al. Novel anti-inflammatory action of edelfosine lacking toxicity with protective effect in experimental colitis. J Pharmacol Exp Ther 2009;329:439–49.

Clinical Cancer Research

277  

 

   

 

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

 

Annex  IV   Lipid  raft-­‐targeted  therapy  in  multiple  myeloma   Faustino  Mollinedo1,  Janis  de  la  Iglesia-­‐Vicente1,  Consuelo  Gajate1,2,  Ander  Estella-­‐ Hermoso  de  Mendoza3,  Janny  A.  Villa-­‐Pulgarin1,  Miguel  A.  Campanero4,  and  Maria  J.   Blanco-­‐Príeto3    

1Centro  de  Investigación  del  Cáncer,  Instituto  de  Biología  Molecular  y  Celular  del  

Cáncer,  CSIC-­‐Universidad  de  Salamanca,  Campus  Miguel  de  Unamuno,  E-­‐37007   Salamanca,  Spain   2Unidad  de  Investigación,  Hospital  Universitario  de  Salamanca,  Campus  Miguel  de  

Unamuno,  E-­‐37007  Salamanca,  Spain  

3Departamento  de  Farmacia  y  Tecnología  Farmacéutica,  Facultad  de  Farmacia,  

Universidad  de  Navarra,  E-­‐31008  Pamplona,  Spain  

4Servicio  de  Farmacología  Clínica,  Clínica  Universitaria,  E-­‐31080  Pamplona,  Spain  

   

 

      Oncogene,  2010.  29(26):  p.  3748-­‐57

279  

 

 

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Oncogene (2010) 29, 3748–3757

& 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 www.nature.com/onc

ORIGINAL ARTICLE

Lipid raft-targeted therapy in multiple myeloma F Mollinedo1, J de la Iglesia-Vicente1, C Gajate1,2, A Estella-Hermoso de Mendoza3, JA Villa-Pulgarin1, MA Campanero4 and MJ Blanco-Prieto3 1

Centro de Investigacio´n del Ca´ncer, Instituto de Biologı´a Molecular y Celular del Ca´ncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain; 2Unidad de Investigacio´n, Hospital Universitario de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain; 3Departamento de Farmacia y Tecnologı´a Farmace´utica, Facultad de Farmacia, Universidad de Navarra, Pamplona, Spain and 4Servicio de Farmacologı´a Clı´nica, Clı´nica Universitaria, Pamplona, Spain

Despite recent advances in treatment, multiple myeloma (MM) remains an incurable malignancy. By using in vitro, ex vivo and in vivo approaches, we have identified here that lipid rafts constitute a new target in MM. We have found that the phospholipid ether edelfosine targets and accumulates in MM cell membrane rafts, inducing apoptosis through co-clustering of rafts and death receptors. Raft disruption by cholesterol depletion inhibited drug uptake by tumor cells as well as cell killing. Cholesterol replenishment restored MM cell ability to take up edelfosine and to undergo drug-induced apoptosis. Ceramide addition displaced cholesterol from rafts, and inhibited edelfosineinduced apoptosis. In an MM animal model, edelfosine oral administration showed a potent in vivo antimyeloma activity, and the drug accumulated preferentially and dramatically in the tumor. A decrease in tumor cell cholesterol, a major raft component, inhibited the in vivo antimyeloma action of edelfosine and reduced drug uptake by the tumor. The results reported here provide the proofof-principle and rationale for further clinical evaluation of edelfosine and for this raft-targeted therapy to improve patient outcome in MM. Our data reveal cholesterolcontaining lipid rafts as a novel and efficient therapeutic target in MM, opening a new avenue in cancer treatment. Oncogene (2010) 29, 3748–3757; doi:10.1038/onc.2010.131; published online 26 April 2010 Keywords: lipid raft; cholesterol; phospholipid ether; edelfosine; apoptosis; MM

Introduction Synthetic alkyl-lysophospholipid analogs (ALPs) constitute a promising family of anticancer drugs that act at Correspondence: Dr F Mollinedo, Centro de Investigacio´n del Ca´ncer, Instituto de Biologı´ a Molecular y Celular del Ca´ncer, CSICUniversidad de Salamanca, Campus Miguel de Unamuno, Salamanca E-37007, Spain. E-mail: [email protected] or Dr C Gajate, Unidad de Investigacio´n, Hospital Universitario de Salamanca, Campus Miguel de Unamuno, Salamanca E-37007, Spain. E-mail: [email protected] Received 23 October 2009; revised 12 January 2010; accepted 7 February 2010; published online 26 April 2010

the membrane level (Gajate and Mollinedo, 2002; Mollinedo et al., 2004; Mollinedo, 2007). The ALP prototype edelfosine (1-O-octadecyl-2-O-methyl-racglycero-3-phosphocholine, ET-18-OCH3) induces apoptosis in tumor cells (Mollinedo et al., 1997) through co-capping of the Fas/CD95 death receptor with membrane rafts (Gajate et al., 2000a, 2004; Gajate and Mollinedo, 2001, 2007). In vitro studies have shown that this phospholipid ether is preferentially taken up by tumor cells, leading to the intracellular activation of the Fas/CD95 death receptor, independently of its ligand FasL/CD95L (Mollinedo et al., 1997; Gajate et al., 2000a, 2004; Gajate and Mollinedo, 2001). Edelfosine-induced apoptosis is mediated by the recruitment of Fas/CD95 and downstream signaling molecules into membrane rafts (Gajate and Mollinedo, 2001, 2007; Gajate et al., 2004). Lipid rafts are membrane microdomains highly enriched in cholesterol and sphingolipids, and the proteins located in these microdomains are limited in their ability to freely diffuse over the plasma membrane, affecting protein function (Simons and Toomre, 2000). Recent evidence suggests that membrane rafts constitute the linchpin from which Fas/CD95 signaling is launched (Gajate et al., 2004, 2009a; Gajate and Mollinedo, 2005, 2007). Multiple myeloma (MM) is the second most prevalent blood cancer, accounting for 10% of all hematological malignancies (Hussein et al., 2002). MM is characterized by the accumulation of malignant plasma cells in the bone marrow with frequent occurrence of lytic bone disease. Although therapeutic advances in the treatment of MM have improved remission duration and overall survival, relapse of the disease is inevitable for nearly all patients, and MM remains a deadly B-cell malignancy (Mihelic et al., 2007). Therefore, development of novel therapeutic strategies is urgently needed to improve survival in MM patients. MM cells show a relatively low proliferative index and a poor apoptotic rate (Dimberg et al., 2005), and hence it is likely that the transforming event is a failure in death regulation rather than a loss of growth control. This implies that a therapeutic potential for MM may lie in potentiating apoptosis, and thereby we hypothesized that MM might be suitable for the particular proapoptotic features of edelfosine. Our previous in vitro data indicated that edelfosineinduced apoptosis in cancer cells, including MM, was mediated by the reorganization of raft protein

  281  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

3749

Results Edelfosine induces Fas/CD95-membrane raft co-clustering and selective killing in MM patientderived cells, sparing normal cells We have previously reported that edelfosine induced apoptosis in human MM cell lines, mediated by coclustering of the Fas/CD95 death receptor in lipid rafts (Gajate and Mollinedo, 2007). Here, we found that edelfosine also induced apoptosis and co-aggregation of Fas/CD95 and rafts in CD138 þ malignant cells derived from MM patients (Figures 1a and b), whereas CD138" normal cells from the same patients, and human normal resting peripheral blood lymphocytes (PBLs), isolated from healthy volunteers, were spared (Figure 1a). MM patient-derived cells took up significant amounts of edelfosine (392±43 pmol per 106 cells after 1 h of incubation with 10 nmol [3H]edelfosine; n ¼ 5), whereas CD138" normal cells from the same patients and normal PBLs did not incorporate the drug (o20 pmol per 106 cells after 1 h of incubation with 10 nmol [3H]edelfosine; n ¼ 3). In addition, normal human cells with distinct proliferation rates, such as purified B cells, human umbilical vein endothelial cells, hepatocytes, CD34 þ hematopoietic progenitor cells and adipose-derived stem cells, were resistant to edelfosine (o3% apoptosis following treatment with 10 or 20 mM edelfosine for 24 h, n ¼ 3) and did not incorporate significant amounts of drug (o20 pmol per 106 cells after 1 h of incubation with 10 nmol [3H]edelfosine; n ¼ 3). By using the raft marker fluorescein isothiocyanate (FITC)-labeled cholera toxin (CTx) B subunit that binds to ganglioside GM1 (Schon and Freire, 1989), mainly found in rafts (Harder et al., 1998), we observed that 10 mM edelfosine promoted a potent co-aggregation of Fas/CD95 and rafts in malignant cells derived from MM patients (Figure 1b). However, no co-clustering of Fas/CD95 and rafts was detected in normal CD138" cells, resting PBLs or purified B cells following incubation with the phospholipid ether (data not shown). Furthermore, incubation of MM patient-derived cells with the cholesterol-depleting agent MCD (Christian et al., 1997) disrupted rafts (Gajate and Mollinedo, 2001), as well as inhibited clustering of Fas/CD95 (data not shown) and apoptosis (41±5 vs 12±3 apoptosis in

a

Control

50

EDLF

% Apoptosis

composition (Gajate and Mollinedo, 2001, 2007; Gajate et al., 2004, 2009a, b), but here we studied whether edelfosine was acting by directly targeting rafts both in vitro and in vivo, using a clinically relevant disease. By using in vitro, ex vivo and in vivo approaches, we show here that edelfosine targets tumor cell lipid rafts and accumulates in these membrane domains, inducing selective apoptosis in MM cells through its preferential uptake in tumor cell rafts. Our data set raft-targeted therapy as a new framework in MM treatment, and identified edelfosine as the first raft-targeted antitumor drug, thus opening a new avenue in cancer chemotherapy.

40 30 20 10 0

b

CD138Raft

CD138+ Fas/CD95

PBLs Merge

Control

Edelfosine

Figure 1 Selective edelfosine-induced apoptosis and Fas/CD95 raft clusters in patient-derived primary MM cells. (a) Freshly isolated CD138 þ tumor MM cells and normal CD138" mononuclear cells from MM patients, as well as normal resting PBLs, were incubated for 24 h with 10 mM edelfosine (EDLF) and analyzed for apoptosis, assessed by the percentage of cells in the sub-G1 region following cell-cycle analysis by flow cytometry. Untreated control cells were run in parallel. Data shown are means±s.e. of five independent determinations. (b) Freshly isolated MM cells from patients were either untreated (control) or treated with 10 mM edelfosine for 9 h, and then stained with FITC-CTx B subunit to identify lipid rafts (green fluorescence) and with a specific anti-Fas/CD95 monoclonal antibody, followed by CY3-conjugated anti-mouse Ig antibody (red fluorescence). Areas of colocalization between membrane rafts and Fas/CD95 in the merge panels are yellow. Images shown are representative of three independent experiments. Bar, 8 mm.

cells untreated vs pretreated with MCD, and then incubated with 10 mM edelfosine for 24 h; n ¼ 3). Thus, the patient-derived MM cells behaved similarly to MM cell lines (Gajate and Mollinedo, 2007) in their capacity to undergo co-clustering of Fas/CD95 rafts as the initial trigger for apoptosis (Mollinedo and Gajate, 2006b; Mollinedo, 2008) on edelfosine treatment. These data suggest that edelfosine shows a remarkable selectivity for primary tumor cells and cancer cell lines in its capacity to promote co-aggregation of Fas/CD95 and rafts and to induce apoptosis. Accumulation of edelfosine in lipid raft clusters enriched in death receptors is critical for MM cell killing We next asked whether edelfosine interacted with MM cell membrane rafts. We tested this directly by Oncogene

  282  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

3750

incubating the MM cell line MM144 with [3H]-edelfosine followed by raft isolation. [3H]-edelfosine accumulated in GM1-containing rafts (Figure 2a). In addition, we found that the fluorescent analog all-[E]-1-O-[150 phenylpentadeca-80 ,100 ,120 ,140 -tetraenyl]-2-O-methyl-racglycero-3-phosphocholine (PTE-edelfosine) (Gajate et al., 2004; Quesada et al., 2004; Nieto-Miguel et al., 2006) accumulated in well-defined patches that colocalized with raft clusters enriched in Fas/CD95 and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors, death receptor (DR) 4 and DR5 (Figure 2b). PTEedelfosine induced apoptosis in MM144 cells (38.6±4.1 and 59.8±5.2% apoptosis following 24-h incubation with 10 and 20 mM PTE-edelfosine, respectively; n ¼ 3) at a similar concentration range as the parent drug edelfosine (51.9±4.3 and 65.6±6.4% apoptosis following 24-h incubation with 10 and 20 mM edelfosine, respectively; n ¼ 3), while sparing normal resting PBLs (o3% apoptosis following 24-h incubation with 10 or 20 mM PTE-edelfosine). Fluorescent PTE-edelfosine incorporation into the MM cell was blocked by addition of the parent drug (data not shown), indicating the specificity in the uptake, and PTE-edelfosine behaves as a bona fide analog to analyze edelfosine localization in the cell. Raft disruption by MCD treatment (Christian et al.,

Fraction

1997) inhibited edelfosine-induced apoptosis (Figure 2c) and clustering of Fas/CD95 and TRAIL receptors (data not shown) in MM144 cells. Similar results were obtained with the MM cell line MM1S (data not shown). These results indicate that edelfosine binds to rafts forming aggregates of edelfosine-rich rafts that recruit death receptors and convey apoptotic signaling. Cholesterol is required for edelfosine uptake and drug-induced apoptosis We found that MM1S and MM144 cells took up edelfosine in the range 415–482 pmol per 106 cells after 1 h of incubation with 10 nmol [3H]edelfosine. To assess the role of cholesterol, a critical raft component, in mediating drug uptake and edelfosine-induced apoptosis, we treated MM cell lines with the cholesterol-depleting agent MCD (Christian et al., 1997). Measurements of [3H]cholesterol levels in cells that had been pre-equilibrated with this lipid showed that treatment with 2.5 mg/ml MCD for 30 min removed >67% of the cell cholesterol, in agreement with previous estimates (Barnes et al., 2004; Gajate et al., 2009b). Disruption of lipid rafts by MCD inhibited both drug uptake (Figure 3a) and edelfosine-induced apoptosis

Raft

Fas/CD95

PTE-EDLF

DIC

Raft

DR4

PTE-EDLF

DIC

Raft

DR5

PTE-EDLF

DIC

3H-EDLF (%)

25 20 15 10 5 0

1 2 3 4 5 6 7 8 9 10 11 12 GM1 128

128

Control

EDLF (51.0%)

Relative number of cells

(1.4 %)

0

0

128

0

1023 MCD (2.5 %)

0

1023

0

0

128

0

1023 MCD + EDLF (9.6%)

0

1023

Relative DNA content (PI Fluorescence)

Figure 2 Edelfosine accumulates in membrane rafts from MM cells enriched in death receptors, leading to apoptosis. (a) MM144 cells treated with 10 mM [3H]edelfosine (0.1 mCi) for 15 h were lysed in 1% Triton and fractionated by centrifugation on a discontinuous sucrose density gradient. An equal volume of each collected fraction was counted for radioactivity and subjected to SDS polyacrylamide gel electrophoresis, and the distributions of [3H]edelfosine (upper panel) and GM1-containing rafts (lower panel) (enriched in fractions 3–5) over the gradient fractions are shown. (b) MM144 cells were incubated with 10 mM PTE-edelfosine (PTEEDLF) (blue fluorescence) for 15 h, washed with PBS–2% BSA, and then its colocalization with membrane rafts and death receptors Fas/CD95, DR4 and DR5 was examined using the FITC-CTx B subunit (green fluorescence for rafts) and specific antibodies against the distinct death receptors, followed by CY3-conjugated antibodies (red fluorescence for death receptors). Differential interference contrast (DIC) images are also shown. Bar, 6 mm. (c) MM144 cells were untreated (control), treated with 10 mM edelfosine for 24 h (EDLF), or pretreated with 2.5 mg/ml MCD for 30 min and then incubated for 24 h in culture medium in the absence (MCD) or presence of 10 mM edelfosine (MCD þ EDLF), and apoptosis was analyzed by flow cytometry. The percentage of apoptotic cells, assessed by a DNA content less than G1 (sub-G1), is shown in parentheses in each histogram. The position of the G1 peak is indicated by arrows. Data shown are representative of three separate experiments. Oncogene

  283  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

3751 Control MCD MCD + CHOL

Control C8-Cer

60 40 20

MM1S

MM144 Control MCD CHOL EDLF MCD + EDLF MCD + CHOL + EDLF

70 60 % Apoptosis

60 40 20 0

50 40

EDLF

60

80

% Apoptosis

% EDLF uptake

% EDLF uptake

80

C8-Cer

70

100

100

0

Control

C8-Cer + EDLF

50 40 30 20 10

MM1S

0

MM1S

Figure 4 C8-ceramide inhibits edelfosine uptake and edelfosineinduced apoptosis in MM cells. (a) MM1S cells were untreated (control), or treated with 5 mM C8-ceramide for 30 min (C8-Cer), and then drug uptake was determined after incubation with 10 mM [3H]edelfosine (0.1 mCi) for 1 h. Data are shown as the percentage of edelfosine uptake, considering the drug uptake in untreated control cells as 100%. (b) MM1S cells were untreated (control), treated with 10 mM edelfosine for 24 h (EDLF), or treated with 5 mM C8-ceramide for 30 min, and then incubated for 24 h in culture medium in the absence (C8-Cer) or presence of 10 mM edelfosine (C8-Cer þ EDLF), and apoptosis was analyzed by flow cytometry as the percentage of hypodiploid (sub-G1) cells following cell-cycle analysis. Data shown are means±s.e. of three independent determinations.

30 20 10 0

MM1S

MM144

Figure 3 Involvement of cholesterol-containing rafts in edelfosine uptake and edelfosine-induced apoptosis in MM cells. (a) MM1S and MM144 cells were untreated (control) or treated with 2.5 mg/ ml MCD for 30 min (MCD), and then drug uptake was determined after incubation with 10 mM [3H]edelfosine (0.1 mCi) for 1 h. Cells were also treated with 2.5 mg/ml MCD for 30 min, followed by cholesterol repletion (MCD þ CHOL) and then drug uptake was examined as above. Data are shown as the percentage of edelfosine uptake, considering the drug uptake in untreated control cells as 100%. (b) MM1S and MM144 cells were untreated (control) and treated with MCD or MCD þ CHOL as above, and then incubated for 24 h in the presence of 10 mM edelfosine (EDLF). Apoptosis was analyzed by flow cytometry as the percentage of hypodiploid (subG1) cells following cell-cycle analysis. Data shown are means±s.e. of three independent determinations.

(Figure 3b). Replenishment of cholesterol cell content by exposure of MCD-treated cells to 100 mg/ml cholesterol for 1 h led to the recovery of B109% cell cholesterol, thus reconstituting cellular cholesterol levels, and cells regained the capacity to take up edelfosine and underwent apoptosis following edelfosine treatment (Figures 3a and b). These results indicate that the presence of cholesterol in rafts is essential for the uptake and apoptotic action of edelfosine. Ceramide inhibits edelfosine-induced apoptosis N-octanoyl-sphingosine (C8-ceramide) is a cell-permeable analog of naturally occurring ceramides that closely

mimics the effects of physiological long-chain ceramide (Karasavvas et al., 1996). Previous reports have shown that both natural and synthetic ceramides specifically displace sterols from lipid rafts affecting their properties and molecular composition (Megha and London, 2004). In this regard, cholesterol is replaced by ceramide in cholesterol-containing rafts to form ceramide-rich rafts (Megha and London, 2004). C8-ceramide has been reported to displace cholesterol from raft domains and destabilize them (Nybond et al., 2005; RouquetteJazdanian et al., 2007). We found that the pretreatment of MM1S cells with C8-ceramide removed cholesterol from the rafts (31±5 vs 9±2 mg cholesterol per mg protein in untreated vs C8-ceramide-treated cells; n ¼ 3), and inhibited both edelfosine uptake (Figure 4a) and edelfosine-induced apoptosis (Figure 4b). These results are in agreement with a critical requirement of cholesterol in rafts for the uptake of edelfosine into MM cells and the subsequent drug-induced apoptotic response. Edelfosine accumulates in MM tumor and inhibits human MM cell growth in vivo We next determined the in vivo antimyeloma activity of edelfosine using a severe combined immune deficiency (SCID) mouse plasmacytoma model. Following toxicity analyses (data not shown), we found that a daily oral administration dose of 30 mg/kg edelfosine was well tolerated, 45 mg/kg being the maximum tolerated dose. CB17-SCID mice were inoculated with 3 " 107 MM1S cells in 100 ml phosphate-buffered saline (PBS), together with 100 ml Matrigel. On the development of a palpable tumor, mice were randomized into drug-treated Oncogene

  284  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

Control

3000 2000 1000

6 5 4 3 2 1 0

EDLF 3 5 9 11 13 16 18 20 23 25 27 30 days

**

Control

EDLF

6 5 4 3 2 1 0

**

Control

Tissue/plasma ratio

0

Tumor weight (g)

Control EDLF

4000

Tumor volume (cm3)

Tumor volume (mm3)

3752

EDLF

**

20 15 10 5 0

Liver

Kidney

Tumor

Figure 5 Edelfosine inhibits human MM cell growth in vivo. CB17-SCID mice were inoculated subcutaneously with 3 " 107 MM1S cells. Daily oral administration of edelfosine (30 mg/kg) started after the development of a palpable tumor. Caliper measurements of longest and shortest diameters of each tumor were carried out at the indicated times. Edelfosine (EDLF) significantly (Po0.01; from day 11 of treatment until the end of the experiment) inhibited MM tumor growth compared with the vehicle-treated control group (a). Data shown in panel a are means±s.e. (n ¼ 12). (b) A remarkable MM tumor growth inhibition was observed after 1-month treatment with edelfosine. Representative tumors isolated from two drug-free MM-bearing mice (control, upper panel) and from three drug-treated MM-bearing mice (EDLF, lower panel) are shown. (c) After completion of the in vivo assay (30 days), drug-free control and edelfosine-treated (EDLF) mice were killed and tumor weight and volume were measured. The tumor size and weight values of each single animal (dots) and the average values of each experimental group (horizontal bars) are shown. **Indicate values that are significantly different at Po0.01. (d) Tissue/plasma concentration ratios, in the liver, kidney and tumor, of edelfosine after daily oral administration of edelfosine (30 mg/kg) for 1 month in MM-bearing SCID mice (means±s.e., n ¼ 8). **Indicate values that are significantly different from liver/plasma and kidney/plasma ratios at P o0.01.

(30 mg/kg edelfosine, daily oral administration for 29 days) and control (water vehicle) groups of 12 mice each. Serial caliper measurements were made to determine the rate at which the tumor grew (Figure 5a). A comparison of tumors isolated from nontreated control and drugtreated MM-bearing mice, at the end of the 1-month treatment, rendered a remarkable and statistically significant (Po0.01) antimyeloma activity of edelfosine, with a reduction of B88% in tumor weight and volume, and four tumor-free mice (Figures 5b and c). Organ examination at necropsy did not reveal any apparent toxicity (data not shown). Compared with the large, highly vascularized tumors from drug-free mice, the small tumors from edelfosine-treated mice were pale and poorly vascularized (Figure 5b). No significant differences in mean body weight were observed between drug-treated and drug-free control animals (3–5% of body weight loss in the treated group vs control group). At the end of the in vivo assay, we found a mean concentration of edelfosine in plasma of 5.42±0.49 mg/ml (n ¼ 12), using high-performance liquid chromatography–mass spectrometry (HPLC–MS) analysis. Interestingly, edelfosine was remarkably more concentrated in the MM tumor (136.83±18.41 mg/g, n ¼ 8) than in the liver (7.91±1.04 mg/g, n ¼ 8) and kidney (13.46±0.26 mg/g, n ¼ 8). Tumor/plasma concentration ratio of edelfosine was dramatically higher than that detected in both kidney and liver, organs that are directly implicated in drug excretion (Figure 5d). Thus, our drug biodistribution studies indicate a rather selective accumulation of edelfosine in the MM tumor in vivo.

Tumor cholesterol levels affect edelfosine antimyeloma activity in vivo To analyze the putative role of rafts in edelfosine action in vivo, the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor pravastatin was given to mice (1.5 mg/kg) to reduce cholesterol levels. An in vivo MM-bearing CB17SCID mouse model assay was conducted involving four groups: untreated control mice, pravastatin-treated mice, edelfosine-treated mice, and mice treated with pravastatin and edelfosine. Pravastatin has been reported to reduce de novo cholesterol synthesis in the liver and in a number of tissues (Koga et al., 1990; Elahi et al., 2008). MM tumors derived from pravastatintreated mice contained a lower content of cholesterol than tumors isolated from untreated mice, 6.9 vs 13.4 mg cholesterol per g of tumor, respectively. Pravastatin significantly inhibited the in vivo antitumor action of edelfosine (Figures 6a, c and d) and drastically decreased the tumor/plasma concentration ratio of edelfosine (Figure 6b). This suggests that tumor cholesterol content has a major role in edelfosine uptake and in the antimyeloma activity of edelfosine in vivo. Although statins are lipid-lowering drugs that block cholesterol biosynthesis, they exert a wide array of effects by inhibiting prenylation of proteins involved in signal transduction (Perez-Sala and Mollinedo, 1994; Perez-Sala et al., 1995; Auer et al., 2002; Jasinska et al., 2007). On these grounds, we cannot rule out the possibility that the above effect of pravastatin could be due, at least in part, to prenylation inhibition.

Oncogene

  285  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

4000 3500 3000 2500 2000 1500 1000 500 0

2

9

23

30 days

2 1 0

20

10 5 0

Liver

EDLF

Pravastatin Pravastatin + EDLF

Tumor

4 3 2 1 0

Control

Kidney

*

*

3

**

15

Tumor volume (cm3)

Tumor weight (g)

4

16

EDLF Pravastatin+EDLF

25 Tissue/plasma ratio

Tumor volume (mm3)

3753 Control EDLF Pravastatin Pravastatin + EDLF

Control

EDLF Pravastatin Pravastatin + EDLF

Figure 6 Pravastatin inhibits the in vivo antitumor action of edelfosine. MM1S-bearing CB17-SCID mice were randomly assigned to four cohorts of eight mice, each receiving a daily oral administration of water vehicle (control), 30 mg/kg edelfosine (EDLF), 1.5 mg/kg pravastatin and pravastatin þ EDLF for 29 days. Tumor shortest and longest diameters were measured weekly to calculate the apparent tumor volume in the above four experimental groups (a). Data shown in panel a are means±s.e. of eight mice in each group. (b) Tissue/plasma concentration ratios, in the liver, kidney and tumor, of edelfosine content after daily oral administration of edelfosine (EDLF) and pravastatin þ EDLF for 1 month in MM-bearing SCID mice (means±s.e., n ¼ 5). **Indicate that the tumor/ plasma ratio values in EDLF and pravastatin þ EDLF are significantly different at Po0.01. Mice were killed 30 days after the initiation of drug treatment, and tumor weight (c) and volume (d) were measured, showing that pravastatin hampered the antitumor effect of edelfosine. *Denotes values that are significantly different between the indicated groups at Po0.05.

Discussion The in vitro and in vivo data reported here identify lipid rafts as a novel and effective therapeutic target in MM, setting a novel framework in cancer chemotherapy. Our results show edelfosine as the first raft-targeted drug, accumulating in lipid rafts and triggering the demise of MM cells. Hence, edelfosine can be considered as the paradigm of this new raft-targeted therapy. The accumulation of edelfosine in rafts might be explained by its high affinity for cholesterol (Ausili et al., 2008; Busto et al., 2008), because of geometry compensation of the ‘cone shape’ of sterols and the ‘inverted cone shape’ of edelfosine that leads to a stable bilayer (Busto et al., 2008). Edelfosine targets membrane rafts of malignant cells, inducing raft aggregates that act as scaffolds for the recruitment and concentration of Fas/ CD95 and TRAIL receptors. Previous reports have shown that MM cells express Fas/CD95, but are rather resistant to undergo apoptosis in response to the external stimulation of Fas/CD95 by agonistic antiFas/CD95 antibodies (Dimberg et al., 2005). However, edelfosine activates Fas/CD95 and induces its aggregation in rafts from within the cell independently of its ligand (Gajate et al., 2004), triggering subsequently downstream extrinsic and intrinsic signaling pathways that eventually lead to apoptosis (Gajate et al., 2000a, b, 2004, 2009a, b; Gajate and Mollinedo, 2001, 2007).

The results reported here, together with our previous findings (Gajate and Mollinedo, 2007; Gajate et al., 2009b), suggest the following cascade of events in edelfosine-induced apoptosis in MM cells: drug accumulation in lipid raft - raft reorganization Fas/CD95 activation - DISC - mitochondrial cytochrome c release - apoptosome. The initial accumulation of edelfosine in lipid rafts is critical to trigger the whole apoptotic pathway, and raft disruption prevents edelfosine uptake and the subsequent activation of both extrinsic and intrinsic pathways (Gajate and Mollinedo, 2007; Gajate et al., 2009b). Although there is increasing evidence that Fas/CD95 apoptotic signaling is initiated through the recruitment of Fas/CD95 receptor in lipid rafts (Mollinedo and Gajate, 2006a, b; Gajate and Mollinedo, 2001, 2007; Mollinedo, 2008), it is not clear whether these domains, initially enriched in cholesterol, must turn into ceramide-rich rafts to deliver apoptotic signals (Grassme et al., 2003). In this regard, edelfosine does not activate sphingomyelinase and no ceramide is generated on its action (Gajate et al., 2004), suggesting that edelfosine acts through cholesterol-rich rafts. As a matter of fact, we found here that the presence of ceramide, which is known to displace cholesterol from lipid rafts (Megha and London, 2004), removes cholesterol from MM cell lipid rafts and inhibits both edelfosine uptake and edelfosine-induced apoptosis. On these grounds, we Oncogene

  286  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

3754

conclude that the lipid rafts involved in both the uptake and apoptotic action of edelfosine are cholesterol-rich rafts and not ceramide-rich rafts. Our biodistribution data in the MM murine model showed a mean concentration of edelfosine in plasma of 5.42 mg/ml (10.35 mM; edelfosine molecular mass, 523.7). Thus, the in vitro effects reported herein, rendered by 10 mM edelfosine, were detected at a pharmacologically relevant concentration. The edelfosine-induced death receptor concentration in specific domains of the cell surface reported here has interesting clinical consequences, as it might either induce apoptosis by itself, through subsequent recruitment of downstream signaling molecules (Gajate and Mollinedo, 2001, 2005, 2007; Gajate et al., 2004, 2009a; Mollinedo and Gajate, 2006a, b), or sensitize cancer cells to death receptor ligands, including TRAIL (Gajate and Mollinedo, 2007), currently under clinical trials. Using an MM xenograft mouse model, we have found that edelfosine accumulates in high amounts in the tumor, highlighting the rather selective affinity of edelfosine for tumor cells. In addition, edelfosine shows a remarkable in vivo antimyeloma activity, leading to drastic tumor regression. Following edelfosine oral administration in nontumor-bearing SCID mice, we have found a rather wide drug distribution pattern to several tissues, including the lung, spleen, intestine, liver and kidney (de Mendoza et al., 2009). Interestingly, we found here that when SCID mice contained MM tumors, edelfosine distribution was dramatically shifted toward the tumor (tumor/plasma concentration ratio D20), indicating a rather selective tumor targeting for edelfosine. We did not find any apparent damage in the distinct organs analyzed following necropsy in the in vivo studies reported here, and have recently found that edelfosine lacked toxicity in rats using histological, functional and biochemical parameters (Mollinedo et al., 2009). Furthermore, the small tumors isolated from edelfosine-treated mice were pale and poorly vascularized. This might be in agreement with reports showing an antiangiogenic effect of edelfosine (Zerp et al., 2008). Interestingly, pharmacological inhibition of endogenous cholesterol synthesis, which diminishes the cholesterol content of membrane rafts (Zhuang et al., 2005), led to a reduction in tumor cholesterol content as well as to a significant decrease in edelfosine antitumor activity and drug accumulation in the tumor. A decrease in cholesterol level might affect lipid rafts, thus hampering edelfosine uptake. Indeed, cholesterol depletion in MM cells inhibited both edelfosine uptake and drug-induced apoptosis. However, cholesterol repletion restored the ability of tumor cells to take up edelfosine and to undergo edelfosine-induced apoptosis. These cholesterol depletion/replenishment assays show that lipid rafts are critical for the uptake of edelfosine and for the triggering of apoptosis by this drug. The finding that edelfosine binds to cholesterol-containing rafts is of particular importance as the cholesterol content in tumor cells has been shown to be higher than in normal cells (Dessi et al., 1994; Kolanjiappan et al., 2003;

Freeman and Solomon, 2004; Tosi and Tugnoli, 2005). Furthermore, cancer cells have been reported to display higher levels of cholesterol-rich lipid rafts than their normal counterparts (Li et al., 2006). Edelfosine shows a high affinity for cholesterol, and for cholesterol-enriched membranes such as rafts (Ausili et al., 2008; Busto et al., 2008), because of the complementarity of the molecular geometrics of sterols and edelfosine (Busto et al., 2008), and this feature may have interesting biomedical applications. The high cholesterol content in tumor cells, together with the avidity for cholesterol of edelfosine, might contribute in part to the accumulation of edelfosine in tumor cells. Nevertheless, the uptake of edelfosine by tumor cells has been suggested to be mediated by a putative protein transporter that remains to be identified (Mollinedo et al., 1997; Hanson et al., 2003). It might be envisaged that tumor cells could undergo a series of changes in lipids, including cholesterol, and proteins, including transporters, which might putatively affect raft organization in a way that facilitates edelfosine uptake. The results reported here provide the proof-ofprinciple and rationale for further clinical evaluation of edelfosine and for this new raft-targeted therapy to improve patient outcome in MM.

Materials and methods Cell lines and primary cells Detailed information on the MM cell lines, primary tumor cells, and normal primary cells used in this study is included in the Supplementary data. Apoptosis assay Quantitation of apoptotic cells was determined by flow cytometry as the percentage of cells in the sub-G1 region (hypodiploidy) in cell-cycle analysis as previously described (Gajate et al., 2000b). Edelfosine was from INKEYSA (Barcelona, Spain) and APOINTECH (Salamanca, Spain). Confocal microscopy Cells were settled onto poly-L-lysine-coated slides and analyzed with a Zeiss LSM 510 laser scan confocal microscope (Oberkochen, Germany) for membrane raft and Fas/CD95 visualization, using FITC-CTx B subunit (Sigma Chemical, St Louis, MO, USA) and anti-human Fas/CD95 SM1/1 IgG2a mouse monoclonal antibody (Bender Medsystems, Vienna, Austria) followed by CY3-conjugated antimouse antibody (Pharmacia, Uppsala, Sweden) as described in Gajate and Mollinedo (2001). Colocalization assays were analyzed by excitation of both fluorochromes in the same section. Negative controls, lacking the primary antibody or using an irrelevant antibody, showed no staining. Edelfosine localization by fluorescence microscopy Cells were treated for 15 h with 10 mM fluorescent analog PTEedelfosine, provided by AU Acun˜a and F Amat-Guerri (Consejo Superior de Investigaciones Cientı´ ficas, Madrid, Spain), and then incubated with the raft marker FITC-CTx B subunit to label lipid rafts, and with an anti-Fas/CD95 SM1/ 1 monoclonal antibody (Bender Medsystems) or specific monoclonal antibodies against the extracellular domains of

Oncogene

  287  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

3755 DR4 and DR5 (R&D Systems, Minneapolis, MN, USA), followed by the corresponding CY3-conjugated secondary antibodies as previously described (Gajate et al., 2004; Gajate and Mollinedo, 2007). Colocalization of the distinct fluorochromes was analyzed using a fluorescence microscope (Axioplan 2; Carl Zeiss MicroImaging) and a digital camera (ORCA-ER-1394; Hamamatsu, Shizuoka, Japan). Negative controls, lacking the primary antibody or using an irrelevant antibody, showed no staining. Edelfosine uptake Drug uptake was measured as previously described (Mollinedo et al., 1997) after incubating 106 cells with 10 nmol [3H]edelfosine for 1 h in RPMI-1640/10% heat-inactivated fetal bovine serum, and subsequent exhaustive washing (six times) with PBS þ 2% bovine serum albumin to remove the ether lipid loosely bound to the cell surface. Lipid raft isolation Lipid rafts were isolated by using lysis conditions and centrifugation on discontinuous sucrose gradients as previously reported (Gajate and Mollinedo, 2001). The location of GM1-containing lipid rafts was determined by western blotting using CTx B subunit conjugated to horseradish peroxidase as previously described (Cheng et al., 1999). Cholesterol depletion and repletion To deplete cholesterol, 2.5–3 " 105 cells per ml were pretreated with 2.5 mg/ml MCD (Sigma) for 30 min at 37 1C in serum-free medium. Cells were then washed three times and resuspended in complete culture medium before the addition of edelfosine. For measurement of relative cholesterol contents, cells were preincubated at 37 1C for 14 h with 0.5 mCi/ml [3H]cholesterol (Perkin Elmer NEN Dupont, Boston, MA, USA) in cellculture medium (Barnes et al., 2004; Gajate et al., 2009b). After incubation without or with MCD, cells were washed, lysed and the radioactivity present in the lysates was then quantified by liquid-scintillation counting. For cholesterol repletion, 100 mg/ml of cholesterol balanced with MCD (watersoluble cholesterol; Sigma) was added for 1 h, and then the cells were washed and resuspended in culture medium for drug treatment. Cholesterol concentration was determined in cells and isolated membrane rafts as previously described (Hope and Pike, 1996; Zhuang et al., 2005), following lipid extraction with chloroform/methanol/HCl, using the Infinity reagent (Thermo Scientific, Worthing, UK). Xenograft mouse models CB17-SCID mice (Charles River Laboratories, Lyon, France), kept and handled according to institutional guidelines, complying with Spanish legislation under a 12/12 h light/dark cycle at a temperature of 22 1C, received a standard diet and acidified water ad libitum. CB17-SCID mice were inoculated subcutaneously into their lower dorsum with 3 " 107 MM1S cells in 100 ml PBS and 100 ml Matrigel basement membrane matrix (Becton Dickinson). When tumors were palpable, mice were randomly assigned to cohorts of 12 mice each, receiving a daily oral administration of edelfosine (30 mg/kg) or an equal volume of vehicle (water). The shortest and longest diameter of the tumor were measured with calipers at the indicated time intervals, and tumor volume (mm3) was calculated using the following standard formula: (the shortest diameter)2 " (the longest diameter) " 0.5. Animals were killed, according to institutional guidelines, when the diameter of their tumors reached 3–4 cm or when significant toxicity was observed.

Animal body weight and any sign of morbidity were monitored. Drug treatment lasted for 29 days, and mice were killed 24 h after the last drug administration. Then, tumors were extirpated, measured and weighed, and a necropsy analysis involving tumors and distinct organs was carried out. For in vivo analysis of the effect of pravastatin, four cohorts of eight CB17-SCID were assigned, receiving daily oral administration of pravastatin (1.5 mg/kg), edelfosine (30 mg/ kg), pravastatin þ edelfosine or an equal volume of vehicle (water). Pravastatin-treated animals received a 1-week pretreatment of pravastatin. Then, analysis was as above. Cholesterol concentration was determined in isolated tumors using a commercial kit (Calbiochem, San Diego, CA, USA) according to the manufacturer’s instructions. Plasma/tissue extraction procedure for edelfosine biodistribution studies MM-bearing SCID mice were treated with a daily oral administration of edelfosine (30 mg/kg) for 29 days. At 24 h after the last drug oral administration, blood was collected in EDTA surface-coated tubes and then centrifuged at 2000 g for 15 min (4 1C) to collect the plasma (100 ml). Then, the animals were killed and distinct organs and tumors were collected and weighed. Tissues and tumors were homogenized in 1 ml water (pH 7.4) using a Mini-bead Beater (BioSpect Products, Inc, Bartlesville, OK, USA) and centrifuged at 10 000 g for 10 min. Both plasma and tissue supernatants were collected and stored at #80 1C until HPLC–MS analysis was carried out. A 10 mg volume of platelet-activating factor (1 mg/ml), used as internal standard, was added onto 100 ml of plasma or tissue/tumor supernatant. A volume of 190 ml of a mixture of 1% formic acid/methanol was added to precipitate proteins. Samples were vortexed for 1 min and, after centrifugation (20 000 g, 10 min), 25 ml of the supernatant were analyzed by HPLC–MS. Quantitative determination of edelfosine by HPLC–MS analysis The technique used was a slight modification of a previously described method (Blanco-Prieto et al., 2004), and is detailed in the Supplementary data. The range of linear response of this technique was 5–250 ng for plasma samples and 5–685 ng for tissue samples. Edelfosine concentrations (mg/ml and mg/g) were calculated dividing the amount of edelfosine measured in samples by the volume of plasma or the weight of tissue used for the analysis. Statistical analysis All values are expressed as means±s.e. Between-group statistical differences were assessed using Mann–Whitney’s test or Student’s t-test. A P-value of o0.05 was considered statistically significant.

Conflict of interest The authors declare no conflict of interest.

Acknowledgements This work was supported by grants from Ministerio de Ciencia e Innovacio´n (SAF2007-61261, SAF2008-02251, PCT-0901002007-27, RD06/0020/1037 from Red Tema´tica de Investigacio´n Cooperativa en Ca´ncer, Instituto de Salud Carlos III), Oncogene

  288  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

3756 Fondo de Investigacio´n Sanitaria and European Commission (FIS-FEDER 06/0813, PS09/01915), Junta de Castilla y Leo´n (GR15-Experimental Therapeutics and Translational Oncology Program, and Biomedicine Project 2009) and Caja Navarra Foundation, Department of Health of the Navarra

Government (‘Ortiz de Landa´zuri, 2009’ project). CG is supported by the Ramo´n y Cajal Program from the Ministerio de Ciencia e Innovacio´n of Spain. AEHdM is supported by a research grant (BF106.37) from the Department of Education of the Basque Government.

References Auer J, Berent R, Weber T, Eber B. (2002). Clinical significance of pleiotropic effects of statins: lipid reduction and beyond. Curr Med Chem 9: 1831–1850. Ausili A, Torrecillas A, Aranda FJ, Mollinedo F, Gajate C, CorbalanGarcia S et al. (2008). Edelfosine is incorporated into rafts and alters their organization. J Phys Chem B 112: 11643–11654. Barnes K, Ingram JC, Bennett MD, Stewart GW, Baldwin SA. (2004). Methyl-beta-cyclodextrin stimulates glucose uptake in Clone 9 cells: a possible role for lipid rafts. Biochem J 378: 343–351. Blanco-Prieto MJ, Campanero MA, Mollinedo F. (2004). Quantitative determination of the antitumor alkyl ether phospholipid edelfosine by reversed-phase liquid chromatography-electrospray mass spectrometry: application to cell uptake studies and characterization of drug delivery systems. J Chromatogr B Analyt Technol Biomed Life Sci 810: 85–92. Busto JV, del Canto-Jan˜ez E, Gon˜i FM, Mollinedo F, Alonso A. (2008). Combination of the anti-tumour cell ether lipid edelfosine with sterols abolishes haemolytic side effects of the drug. J Chem Biol 1: 89–94. Cheng PC, Dykstra ML, Mitchell RN, Pierce SK. (1999). A role for lipid rafts in B cell antigen receptor signaling and antigen targeting. J Exp Med 190: 1549–1560. Christian AE, Haynes MP, Phillips MC, Rothblat GH. (1997). Use of cyclodextrins for manipulating cellular cholesterol content. J Lipid Res 38: 2264–2272. de Mendoza AE, Campanero MA, de la Iglesia-Vicente J, Gajate C, Mollinedo F, Blanco-Prieto MJ. (2009). Antitumor alkyl ether lipid edelfosine: tissue distribution and pharmacokinetic behavior in healthy and tumor-bearing immunosuppressed mice. Clin Cancer Res 15: 858–864. Dessi S, Batetta B, Pulisci D, Spano O, Anchisi C, Tessitore L et al. (1994). Cholesterol content in tumor tissues is inversely associated with high-density lipoprotein cholesterol in serum in patients with gastrointestinal cancer. Cancer 73: 253–258. Dimberg LY, Dimberg AI, Ivarsson K, Stromberg T, Osterborg A, Nilsson K et al. (2005). Ectopic and IFN-induced expression of Fas overcomes resistance to Fas-mediated apoptosis in multiple myeloma cells. Blood 106: 1346–1354. Elahi MM, Cagampang FR, Anthony FW, Curzen N, Ohri SK, Hanson MA. (2008). Statin treatment in hypercholesterolemic pregnant mice reduces cardiovascular risk factors in their offspring. Hypertension 51: 939–944. Freeman MR, Solomon KR. (2004). Cholesterol and prostate cancer. J Cell Biochem 91: 54–69. Gajate C, Del Canto-Janez E, Acuna AU, Amat-Guerri F, Geijo E, Santos-Beneit AM et al. (2004). Intracellular triggering of Fas aggregation and recruitment of apoptotic molecules into Fas-enriched rafts in selective tumor cell apoptosis. J Exp Med 200: 353–365. Gajate C, Fonteriz RI, Cabaner C, Alvarez-Noves G, AlvarezRodriguez Y, Modolell M et al. (2000a). Intracellular triggering of Fas, independently of FasL, as a new mechanism of antitumor ether lipid-induced apoptosis. Int J Cancer 85: 674–682. Gajate C, Gonzalez-Camacho F, Mollinedo F. (2009a). Involvement of raft aggregates enriched in Fas/CD95 death-inducing signaling complex in the antileukemic action of edelfosine in Jurkat cells. PLoS ONE 4: e5044. Gajate C, Gonzalez-Camacho F, Mollinedo F. (2009b). Lipid raft connection between extrinsic and intrinsic apoptotic pathways. Biochem Biophys Res Commun 380: 780–784.

Gajate C, Mollinedo F. (2001). The antitumor ether lipid ET-18-OCH3 induces apoptosis through translocation and capping of Fas/CD95 into membrane rafts in human leukemic cells. Blood 98: 3860–3863. Gajate C, Mollinedo F. (2002). Biological activities, mechanisms of action and biomedical prospect of the antitumor ether phospholipid ET-18-OCH3 (edelfosine), a proapoptotic agent in tumor cells. Curr Drug Metab 3: 491–525. Gajate C, Mollinedo F. (2005). Cytoskeleton-mediated death receptor and ligand concentration in lipid rafts forms apoptosis-promoting clusters in cancer chemotherapy. J Biol Chem 280: 11641–11647. Gajate C, Mollinedo F. (2007). Edelfosine and perifosine induce selective apoptosis in multiple myeloma by recruitment of death receptors and downstream signaling molecules into lipid rafts. Blood 109: 711–719. Gajate C, Santos-Beneit AM, Macho A, Lazaro M, Hernandez-De Rojas A, Modolell M et al. (2000b). Involvement of mitochondria and caspase-3 in ET-18-OCH3-induced apoptosis of human leukemic cells. Int J Cancer 86: 208–218. Grassme H, Cremesti A, Kolesnick R, Gulbins E. (2003). Ceramidemediated clustering is required for CD95-DISC formation. Oncogene 22: 5457–5470. Hanson PK, Malone L, Birchmore JL, Nichols JW. (2003). Lem3p is essential for the uptake and potency of alkylphosphocholine drugs, edelfosine and miltefosine. J Biol Chem 278: 36041–36050. Harder T, Scheiffele P, Verkade P, Simons K. (1998). Lipid domain structure of the plasma membrane revealed by patching of membrane components. J Cell Biol 141: 929–942. Hope HR, Pike LJ. (1996). Phosphoinositides and phosphoinositideutilizing enzymes in detergent-insoluble lipid domains. Mol Biol Cell 7: 843–851. Hussein MA, Juturi JV, Lieberman I. (2002). Multiple myeloma: present and future. Curr Opin Oncol 14: 31–35. Jasinska M, Owczarek J, Orszulak-Michalak D. (2007). Statins: a new insight into their mechanisms of action and consequent pleiotropic effects. Pharmacol Rep 59: 483–499. Karasavvas N, Erukulla RK, Bittman R, Lockshin R, Zakeri Z. (1996). Stereospecific induction of apoptosis in U937 cells by Noctanoyl-sphingosine stereoisomers and N-octyl-sphingosine. The ceramide amide group is not required for apoptosis. Eur J Biochem 236: 729–737. Koga T, Shimada Y, Kuroda M, Tsujita Y, Hasegawa K, Yamazaki M. (1990). Tissue-selective inhibition of cholesterol synthesis in vivo by pravastatin sodium, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor. Biochim Biophys Acta 1045: 115–120. Kolanjiappan K, Ramachandran CR, Manoharan S. (2003). Biochemical changes in tumor tissues of oral cancer patients. Clin Biochem 36: 61–65. Li YC, Park MJ, Ye SK, Kim CW, Kim YN. (2006). Elevated levels of cholesterol-rich lipid rafts in cancer cells are correlated with apoptosis sensitivity induced by cholesterol-depleting agents. Am J Pathol 168: 1107–1118; quiz 1404-1105. Megha, London E. (2004). Ceramide selectively displaces cholesterol from ordered lipid domains (rafts): implications for lipid raft structure and function. J Biol Chem 279: 9997–10004. Mihelic R, Kaufman JL, Lonial S. (2007). Maintenance therapy in multiple myeloma. Leukemia 21: 1150–1157. Mollinedo F. (2007). Antitumor ether lipids: proapoptotic agents with multiple therapeutic indications. Expert Opin Ther Pat 17: 385–405.

Oncogene

  289  

  Annex  IV.  Lipid  raft-­‐targeted  therapy  in  multiple  myeloma    

Lipid raft-targeted therapy in multiple myeloma F Mollinedo et al

3757 Mollinedo F. (2008). Myeloma Therapy. Pursuing The Plasma Cell. Lonial S (ed). Humana Press: Totowa, NJ, pp Chapter 25, pp. 393–419. Mollinedo F, Fernandez-Luna JL, Gajate C, Martin-Martin B, Benito A, Martinez-Dalmau R et al. (1997). Selective induction of apoptosis in cancer cells by the ether lipid ET-18-OCH3 (edelfosine): molecular structure requirements, cellular uptake, and protection by Bcl-2 and Bcl-XL. Cancer Res 57: 1320–1328. Mollinedo F, Gajate C. (2006a). Fas/CD95 death receptor and lipid rafts: new targets for apoptosis-directed cancer therapy. Drug Resist Updat 9: 51–73. Mollinedo F, Gajate C. (2006b). Fas Signaling. Wajant H (ed). Landes Bioscience and Springer Science: Georgetown, TX, pp Chapter 2, pp 13–27. Mollinedo F, Gajate C, Martin-Santamaria S, Gago F. (2004). ET-18OCH3 (edelfosine): a selective antitumour lipid targeting apoptosis through intracellular activation of Fas/CD95 death receptor. Curr Med Chem 11: 3163–3184. Mollinedo F, Gajate C, Morales AI, del Canto-Janez E, Justies N, Collia F et al. (2009). Novel anti-inflammatory action of edelfosine lacking toxicity with protective effect in experimental colitis. J Pharmacol Exp Ther 329: 439–449. Nieto-Miguel T, Gajate C, Mollinedo F. (2006). Differential targets and subcellular localization of antitumor alkyl-lysophospholipid in leukemic versus solid tumor cells. J Biol Chem 281: 14833–14840. Nybond S, Bjorkqvist YJ, Ramstedt B, Slotte JP. (2005). Acyl chain length affects ceramide action on sterol/sphingomyelin-rich domains. Biochim Biophys Acta 1718: 61–66.

Perez-Sala D, Collado-Escobar D, Mollinedo F. (1995). Intracellular alkalinization suppresses lovastatin-induced apoptosis in HL-60 cells through the inactivation of a pH-dependent endonuclease. J Biol Chem 270: 6235–6242. Perez-Sala D, Mollinedo F. (1994). Inhibition of isoprenoid biosynthesis induces apoptosis in human promyelocytic HL-60 cells. Biochem Biophys Res Commun 199: 1209–1215. Quesada E, Delgado J, Gajate C, Mollinedo F, Acuna AU, AmatGuerri F. (2004). Fluorescent phenylpolyene analogues of the ether phospholipid edelfosine for the selective labeling of cancer cells. J Med Chem 47: 5333–5335. Rouquette-Jazdanian AK, Pelassy C, Breittmayer JP, Aussel C. (2007). Full CD3/TCR activation through cholesterol-depleted lipid rafts. Cell Signal 19: 1404–1418. Schon A, Freire E. (1989). Thermodynamics of intersubunit interactions in cholera toxin upon binding to the oligosaccharide portion of its cell surface receptor, ganglioside GM1. Biochemistry 28: 5019–5024. Simons K, Toomre D. (2000). Lipid rafts and signal transduction. Nat Rev Mol Cell Biol 1: 31–39. Tosi MR, Tugnoli V. (2005). Cholesteryl esters in malignancy. Clin Chim Acta 359: 27–45. Zerp SF, Vink SR, Ruiter GA, Koolwijk P, Peters E, van der Luit AH et al. (2008). Alkylphospholipids inhibit capillary-like endothelial tube formation in vitro: antiangiogenic properties of a new class of antitumor agents. Anticancer Drugs 19: 65–75. Zhuang L, Kim J, Adam RM, Solomon KR, Freeman MR. (2005). Cholesterol targeting alters lipid raft composition and cell survival in prostate cancer cells and xenografts. J Clin Invest 115: 959–968.

Supplementary Information accompanies the paper on the Oncogene website (http://www.nature.com/onc)

Oncogene

  290  

  Annex  V.  Commentary:  The  Rafts  of  the  Medusa:  cholesterol  targeting  in  cancer  therapy    

   

Annex  V    

Commentary:  The  Rafts  of  the  Medusa:  cholesterol   targeting  in  cancer  therapy     MR  Freeman,  D  Di  Vizio  and  KR  Solomon             Oncogene,  2010.  29:  p.  3745-­‐47            

291  

 

     

 

  Annex  V.  Commentary:  The  Rafts  of  the  Medusa:  cholesterol  targeting  in  cancer  therapy    

Oncogene (2010) 29, 3745–3747

& 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 www.nature.com/onc

COMMENTARY

The Rafts of the Medusa: cholesterol targeting in cancer therapy MR Freeman1,2,3,4, D Di Vizio1,2,3 and KR Solomon1,2,5 1 Urological Diseases Research Center, Children’s Hospital Boston, Boston, MA, USA; 2Department of Urology, Children’s Hospital Boston–Harvard Medical School, Boston, MA, USA; 3Department of Surgery, Children’s Hospital Boston–Harvard Medical School, Boston, MA, USA; 4Department of Biological Chemistry and Molecular Pharmacology, Children’s Hospital Boston–Harvard Medical School, Boston, MA, USA and 5Department of Orthopaedic Surgery, Children’s Hospital Boston–Harvard Medical School, Boston, MA, USA

In this issue of Oncogene, Mollinedo and co-workers present promising evidence that cholesterol-sensitive signaling pathways involving lipid rafts can be therapeutically targeted in multiple myeloma. Because the pathways considered in their study are used by other types of tumor cells, one implication of this report is that cholesterol-targeting approaches may be applicable to other malignancies. Oncogene (2010) 29, 3745–3747; doi:10.1038/onc.2010.132; published online 3 May 2010

Cholesterol is a sterol that serves as a metabolic precursor to other bioactive sterols, such as nuclear receptor ligands, and also has a major role in plasma membrane structure. Cholesterol and longchain sphingolipids are believed to accumulate in ‘liquid-ordered’ patches termed lipid rafts, which are distinct in dynamic behavior from cholesterol-poor regions of the membrane. Biophysical, biochemical and imaging studies have provided strong evidence that lipid rafts exist in biological membranes, although details of their precise form and stability are still hotly debated. Cholesterol-rich microdomains have been shown to sequester a variety of membraneassociated signaling proteins, while excluding others, and a variety of approaches have shown that cholesterol itself has a role in transmitting cell growth, survival and differentiation signals. There has been speculation that cholesteroldependent membrane dynamics, such as receptor clustering, may be

Correspondence: Dr MR Freeman, Urological Diseases Research Center, Enders 1161, Children’s Hospital Boston, 300 Longwood Avenue, Boston, MA 02115, USA. E-mail: [email protected] Received 14 March 2010; accepted 14 March 2010; published online 3 May 2010

targeted therapeutically in the case of certain malignancies. Published evidence suggests that a cholesterol-focused approach might work in some clinical scenarios. Cholesterol-lowering drugs that inhibit the enzyme HMG-CoA reductase, which catalyzes the ratelimiting step in cholesterol synthesis (these drugs are generically termed ‘statins’), have been reported to inhibit cancer incidence or progression in some studies. Although there is much controversy, buttressed by claims and counterclaims, in the various population-based reports of the effects of statins on cancer, recent evidence published by several groups examining large prospective series suggest that prostate cancer progression is likely to be inhibited by long-term cholesterol-lowering therapy (Platz et al., 2006; Mondul et al., 2010). These promising results in humans are in agreement with animal model data, in which cholesterol is raised or lowered and prostate tumor growth thereby promoted or inhibited, respectively (Zhuang et al., 2005; Solomon et al., 2009). Prostate cancer may be a special case, however, because recent observations from several groups indicate that tumor cells are capable of de novo androgen synthesis from cholesterol, obviating the need to acquire the hormone from the circulation. Because

androgens are generally thought to promote prostate cancer disease progression, the relative clarity of the epidemiological data in prostate cancer in comparison to other organ sites may arise from the role of cholesterol as the synthetic precursor of androgens. The paper by Mollinedo et al. in this issue, along with previous studies from this group and from other labs, provide persuasive evidence that membrane cholesterol itself is a critical mediator of cell survival signaling mechanisms that can be effectively targeted with clinically relevant drugs. Because this has been shown in multiple myeloma, a malignancy without the hormone dependence of prostate cancer, the in situ production of steroid hormones is an unlikely explanation for the sensitivity to cholesterol-targeting manipulations. Edelfosine, and the related compound, perifosine, are prototypic members of a family of synthetic lipids with tumor-killing properties known as alkyl-lysophospholipid analogs. Both drugs are presently in human trials and significant clinical responses have been reported. In a series of studies, edelfosine was shown to accumulate in cell membrane rafts and cause co-clustering of components of the death-inducing signaling complex, resulting in tumor cell apoptosis.

  293  

  Annex  V.  Commentary:  The  Rafts  of  the  Medusa:  cholesterol  targeting  in  cancer  therapy    

The Rafts of the Medusa MR Freeman et al

3746

Raft disruption by cholesterol depletion, using a variety of distinct approaches, inhibits drug uptake by tumor cells, preventing apoptosis. This inhibition is reverted by cholesterol replenishment. in vivo, edelfosine accumulates in mouse plasmacytomas, inducing apoptosis. However, the drug uptake by the tumor is reduced if tumor cholesterol levels are lowered by statin therapy, resulting in treatment failure. These and supporting observations strongly suggest that tumor apoptosis induced by the alkyllysophospholipid compounds is cholesterol dependent. Edelfosine was the first compound reported to promote apoptosis by ligand-independent activation of Fas/CD95 death receptors. However, this class of drug may also operate by cholesterol-sensitive mechanisms distinct from death-inducing signaling complex formation. Perifosine was shown to potently inhibit activation of the serine-threonine kinase Akt (Hideshima et al., 2006), which usually performs a prosurvival function. In other studies, Akt has been identified as a cholesterol-sensitive protein that localizes to lipid rafts (Adam et al., 2007), consistent with a mechanism of action where perifosine alters raftmediated signaling from Akt. Collectively, these results suggest that cholesterol/lipid raft targeting may cooperatively activate both prodeath and inhibit pro-survival pathways simultaneously. One important issue is that edelfosine is taken up specifically by CD138 þ malignant cells obtained from patients with multiple myeloma, whereas CD138" normal cells from the same patients and normal peripheral blood lymphocytes did not incorporate the drug. Similarly, edelfosine induced clustering of Fas/CD95 and TRAIL receptors in lipid rafts in tumor cells, but not in normal cells from the same patients. In vivo, edelfosine concentrated in the multiple myeloma tumors but the drug localized poorly in liver and kidney. These observations suggest that tumor cell membranes are substantially altered in lipid composition, a consequence of malignant transformation that

could be exploited with lipid-targeting agents. Lipid targeting in cancer therapy is of growing interest because of a reawakened appreciation for the role of metabolic pathways as essential features of malignant transformation and progression (Vander Heiden et al., 2009). Tumor cells consume high levels of glucose but often simultaneously suppress ATP production, diverting carbon atoms and reducing equivalents toward pathways that synthesize macromolecules, particularly lipids required for new membrane formation. Upregulation of fatty acid synthase, the major source of long-chain fatty acids (principally palmitate) in tumor cells, is a prominent characteristic of this metabolic profile (Menendez and Lupu, 2007). Lipogenesis is also activated by Akt upregulation, another common event in malignancy. Preclinical data have shown that fatty acid synthase is effectively targetable in vivo and efforts are underway to improve the toxicity profile associated with the available anti-fatty acid synthase compounds. Inhibiting various components of the growth factor receptor-PI3kinase-Akt-mTOR pathway, an active area in pharmaceutical R&D, will produce major alterations in lipid metabolism, only some of which are known. The landscape of tumor cell membranes is drastically altered from normal as a result of shifts in metabolism arising from malignant transformation; however, characterization of membrane lipid composition in tumor cells is still at a fairly primitive stage. Given the absolute requirement for cholesterol in the synthesis of mammalian cell membranes, it follows that rapidly proliferating tumor cells require more cholesterol than normal cells. Moreover, the ability of cancer cells to metastasize may depend on the formation of cholesterol-rich cell extensions called invadopodia, which may not form in the absence of excess cholesterol (Caldieri and Buccione, 2010). Thus, metastatic cells’ dependence on abnormal levels of cholesterol may prove to be their undoing if vulnerabilities in lipid metabolism can be identified

and exploited. As the tumor metabolism picture becomes clearer over the next decade, lipid-targeting strategies should prove relevant to the clinical situation, at least for some classes of tumors. Because of shared metabolic consequences of cell transformation that arise from the many varieties of genetic alterations in multiple signal transduction pathways, one can even imagine that lipid-targeting approaches may ultimately prove to be more widely efficacious than is currently the case for drugs directed toward the major protein signal transduction targets, such as ErbB family receptors and other kinases. There are some caveats that should be considered when evaluating the fortunes of cholesterol targeting. Modeling cholesterol reduction in rodents is challenging because statin drugs do not lower circulating cholesterol in mice and rats, as they do in humans. Cholesterol lowering in rodent tumors will thus depend on whether the drug can penetrate sufficiently into the tumor bed, a scenario that does not adequately model the conventional cholesterol reduction situation in humans, in which the level of extrahepatic statin is low and of short duration. However, other approaches to cholesterol lowering in mice have recently been developed (Solomon et al., 2009) and these might be exploited to identify cholesterol-sensitive tumor types by way of xenografts or transgenic models. Whether the unusually high level of selectivity toward tumor cells seen with edelfosine will be replicated with other lipid-targeting agents is an open question. In addition, high concentrations of cholesterol are naturally found in the brain, ilium, liver and prostate. Consequently, a cholesterol-targeting approach may not be selective for tumors at certain anatomical sites. It is also not clear whether edelfosine can cross either the blood–brain or blood–testes barriers, so malignancies at these locations may not be targetable by the alkyl-lysophospholipid class of compounds. A not unfitting metaphor for cancer, Medusa was a mythical

Oncogene

  294  

  Annex  V.  Commentary:  The  Rafts  of  the  Medusa:  cholesterol  targeting  in  cancer  therapy    

The Rafts of the Medusa MR Freeman et al

3747

creature, highly feared, with snakes in place of hair, which could turn someone to stone with a single glance. The Raft of the Medusa (Le Radeau de la Me´duse) is a famous oil painting by The´odore Ge´ricault, which depicts a tragic escape from a wrecked French naval vessel of the same name.

Although most of the castaways lost their lives in the escape attempt, some were saved by the hastily constructed raft. It is intriguing that the metaphor of the Raft of the Medusa providing sanctuary and life may someday extend to patients, where the cholesterol- and lipid-rich membrane environment

of tumors might provide effective therapeutic opportunities.

cancer pathogenesis. Nat Rev Cancer 7: 763–777. Mondul AM, Clipp SL, Helzlsouer KJ, Platz EA. (2010). Association between plasma total cholesterol concentration and incident prostate cancer in the CLUE II cohort. Cancer Causes Control 21: 61–68. Mollinedo F, de la Iglesia-Vicente J, Gajate C, Estella-Hermoso de Mendoza A, VillaPulgarin JA, Campanero MA et al. (2010). Lipid raft-targeted therapy in multiple myeloma. Oncogene 29: 3748–3757. Platz EA, Leitzmann MF, Visvanathan K, Rimm EB, Stampfer MJ, Willett WC et al. (2006). Statin drugs and risk of advanced

prostate cancer. J Natl Cancer Inst 98: 1819–1825. Solomon KR, Pelton K, Boucher K, Joo J, Tully C, Zurakowski D et al. (2009). Ezetimibe is an inhibitor of tumor angiogenesis. Am J Pathol 174: 1017–1026. Vander Heiden MG, Cantley LC, Thompson CB. (2009). Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324: 1029–1033. Zhuang L, Kim J, Adam RM, Solomon KR, Freeman MR. (2005). Cholesterol targeting alters lipid raft composition and cell survival in prostate cancer cells and xenografts. J Clin Invest 115: 959–968.

Conflict of interest The authors declare no conflict of interest.

References Adam RM, Mukhopadhyay NK, Kim J, Di Vizio D, Cinar B, Boucher K et al. (2007). Cholesterol sensitivity of endogenous and myristoylated Akt. Cancer Res 67: 6238–6246. Caldieri G, Buccione R. (2010). Aiming for invadopodia: organizing polarized delivery at sites of invasion. Trends Cell Biol 20: 64–70. Hideshima T, Catley L, Yasui H, Ishitsuka K, Raje N, Mitsiades C et al. (2006). Perifosine, an oral bioactive novel alkylphospholipid, inhibits Akt and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells. Blood 107: 4053–4062. Menendez JA, Lupu R. (2007). Fatty acid synthase and the lipogenic phenotype in

Oncogene

  295  

Smile Life

When life gives you a hundred reasons to cry, show life that you have a thousand reasons to smile

Get in touch

© Copyright 2015 - 2024 PDFFOX.COM - All rights reserved.