213
Departamento de Farmacia y Tecnología Farmacéutica Facultad de Farmacia y Nutrición UNIVERSIDAD DE NAVARRA TESIS DOCTORAL Design and in vitro / in vivo evaluation of a targeted immunotherapy platform for the treatment of melanoma“Diseño y evaluación in vitro / in vivo de una plataforma dirigida de inmunoterapia para el tratamiento de melanoma” Trabajo presentado por María Merino Díaz para obtener el Grado de Doctor María Merino Díaz Pamplona, 2018

Design and in vitro / in vivo evaluation of a targeted ...dadun.unav.edu/bitstream/10171/56360/1/Tesis_MerinoDiaz18.pdfComo no a mis amig@s, Sani, Ire, Car, Alex y Cris, sois y seréis

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

  • Departamento de Farmacia y Tecnología Farmacéutica

    Facultad de Farmacia y Nutrición

    UNIVERSIDAD DE NAVARRA

    TESIS DOCTORAL

    “Design and in vitro / in vivo evaluation of a targeted

    immunotherapy platform for the treatment of melanoma”

    “Diseño y evaluación in vitro / in vivo de una plataforma

    dirigida de inmunoterapia para el tratamiento de melanoma”

    Trabajo presentado por María Merino Díaz para obtener el Grado de Doctor

    María Merino Díaz

    Pamplona, 2018

  • UNIVERSIDAD DE NAVARRA

    FACULTAD DE FARMACIA Y NUTRICIÓN

    Departamento de Farmacia y Tecnología Farmacéutica

    Memoria presentada por Dña. María Merino Díaz para aspirar al grado de Doctor por la

    Universidad de Navarra.

    Fdo. María Merino Díaz

    El presente trabajo ha sido realizado bajo nuestra dirección en el Departamento de

    Farmacia y Tecnología Farmacéutica de la Facultad de Farmacia y Nutrición de la

    Universidad de Navarra y autorizamos su presentación ante el Tribunal que lo ha de juzgar.

    VºBº Director VºBº Co-Director

    María Jesús Garrido Cid Sara Zalba Oteiza

  • Las investigaciones realizadas en el presente trabajo se han

    desarrollado dentro del proyecto “Estudio de agentes

    inmunomoduladores en oncología mediante plataformas preclínicas

    para desarrollar un modelo cinético-dinámico con carácter

    traslacional” financiado por el Plan de Investigación de la

    Universidad de Navarra (PIUNA).

  • A mi familia y Enrique.

  • AGRADECIMIENTOS

    AGRADECIMIENTOS

    En primer lugar me gustaría expresar mi agradecimiento a la Universidad de

    Navarra y al Departamento de Farmacia y Tecnología Farmacéutica por haberme permitido

    realizar esta tesis doctoral.

    A la Dra. María Jesús Garrido Cid y al Dr. Ignacio Fernández Trocóniz, por

    haberme dado la oportunidad de realizar la tesis en su grupo. En especial a María Jesús,

    muchas gracias por la dirección de esta tesis, por guiarme y aconsejarme en estos años, y

    por la confianza puesta en mí y en mi trabajo.

    A los investigadores, profesores y personal del Departamento de Farmacia y

    Tecnología Farmacéutica: Socorro Espuelas, Carmen Dios, Maribel Calvo, María del Mar

    Goñi, Juan Manuel Irache, Elisa Garbayo, Conchita Tros, Fernando Martínez, Félix

    Recarte, María Huici, Noelia Ruz y María José Blanco, por compartir día a día el lugar de

    trabajo y por la ayuda prestada cuando así lo he necesitado. A Hugo Lana, gracias por tu

    ayuda y por hacerme valorar el trabajo bien hecho.

    A los compañeros del departamento, los que están y los que ya se han ido, Nekane,

    Simón, Laura S, Carlos, Meli, Cristina, Paula, Laura I, Juana, Koldo, Leire, Itziar, Violeta,

    Zinnia, Edu, Ana, JD, María G, Belén, Víctor, Nuria, Nacho, Diego, por compartir tantas

    experiencias de trabajo, aprendizajes y ratos agradables a lo largo de estos años.

    A todos los que han hecho posible que este trabajo saliera adelante: al departamento

    de Microbiología y al departamento de Histología y Anatomía Patológica de la

    Universidad de Navarra, a la Unidad de Imagen y Morfología del CIMA.

    My most sincere gratitude to all the people of the Erasmus Medical Center of

    Rotterdam, The Netherlands, for their hospitality and support during my stay there. In

    particular to Timo L.M. ten Hagen, thank you so much for accepting me going to your lab,

    giving me advice and for the confidence you placed on me. I learnt a lot working there.

    Gracias a toda la gente del CIMA, Noelia, Teresa, Juanjo y Sandra, y en particular

    al laboratorio 3.03, Marcos, Celia, Nuria y Kepa. Gracias por todos esos cafés y charlas y

    por la ayuda y el apoyo que me habéis prestado durante todos estos años.

    A las chicas del coche, Ana, Miriam y Sara G. Pero un especial cariño a Inma y a

    Marina, por esas largas charlas existenciales y el apoyo, compresión y cariño que habéis

  • AGRADECIMIENTOS

    mostrado todos estos años. ¡¡Sin vosotras los viajes diarios habrían sido interminables y os

    habéis convertido en verdaderas amigas!!

    A mis chihuahuas, Lina, Esther, Yolanda, Alba, Inés, Ana Luisa, Edurne L, Jorge y

    Cristian, millones de gracias por los momentos que hemos vivido todos estos años. Por

    esas comidas y excursiones, por apoyarme y ayudarme a intentar encontrar opciones y

    salidas a tantos problemas. Pero sobre todo por vuestra amistad, por hacerme sonreír,

    animarme y hacerme ver el vaso lleno cuando lo veía completamente vacío.

    A Ana M y Laura B, qué diría de vosotras. Me habéis acompañado en toda esta

    tesis. Millones de gracias por todo este tiempo, por lo que me habéis enseñado, vuestra

    ayuda, apoyo, cariño y compresión. ¡¡Gracias por vuestra amistad!!

    A Sara, qué decir de Sara Zarrrba. Me metiste en esto, y tú me has sacado…Gracias

    por todos estos años, por tu paciencia, por enseñarme, por hacerme ver este mundo de la

    forma que tú lo ves, por tu amistad, apoyo, ánimos y sinceridad. Sara ya sabes que sin tu

    ayuda esta tesis no habría salido adelante, así que ¡¡GRACIAS POR TODO!!

    A todo el AJN, en especial a Iñi, Miguel y Javi, por taaaantos y taaaantos

    momentos juntos, ¡y los que nos quedan!. Pero sobretodo y con muchísimo cariño a mis

    Belenchu y Alber, gracias por aguantarme incluso sin comprender lo que decía, gracias por

    todos los viajes juntos, vacaciones, salidas, excursiones, cenas…¡¡Sois geniales!!

    A la familia Buil-Herreros de Tejada, y muy especial a Pepa, Juan, Pauli y David.

    Gracias por el apoyo que me habéis dado desde que os conocí.

    Como no a mis amig@s, Sani, Ire, Car, Alex y Cris, sois y seréis los mejores

    amigos que se puede tener. Gracias por haber estado ahí en todo momento, por haber

    aguantado tantos y tantos audios y conversaciones sobre algo que ni entendíais, por

    apoyarme en esta larga aventura y darme ánimos y esperanza. Gracias por hacerme ver el

    lado positivo de las cosas y hacerme reír en momento difíciles. ¡¡¡Sois l@s mejores!!!

    A mi familia, en particular a mis yayos. Habéis sido, sois y seréis una parte muy

    importante de mi vida, me habéis apoyado en todo lo que he decidido hacer sin poner en

    duda mi capacidad para ello, siempre alegres y contentos de poder pasar ratos juntos,

    aunque ésta tesis me haya hecho perder algunos. Pase lo que pase SIEMPRE os voy a

    querer.

    Mis últimas palabras son para las personas más importantes de mi vida, mis padres.

    Nunca os podré agradecer todo lo que habéis hecho y estáis haciendo por mí, por la

    educación que me habéis dado, y los valores que me habéis sabido transmitir. Sin vosotros

  • AGRADECIMIENTOS

    y vuestro apoyo incondicional no habría podido hacer muchas de las cosas que he hecho.

    ¡¡OS QUIERO!!

    Y por último Enrique…qué decir de Enrique. Gracias por esas visitas a Italia,

    Inglaterra y Holanda. Gracias por tu paciencia, tu amabilidad, tu comprensión y tu cariño.

    Gracias por saber estar ahí en todo momento intentando sacarme una sonrisa. Gracias por

    haber aguantado todos los momentos de estrés y haberme apoyado en ésta aventura ¡Nos

    vemos el 29 de Junio!

    A todos los que de alguna forma han formado parte de esta aventura,

    ¡MUCHISIMAS GRACIAS!

  • CONTENTS

    CONTENTS

    ABBREVIATIONS ............................................................................................................... 1

    INTRODUCTION ................................................................................................................ 5

    References ...................................................................................................................... 13

    CHAPTER 1

    Immunoliposomes in clinical oncology: State of the art and future

    perspectives

    Abstract ............................................................................................................................ 21

    1. Introduction .................................................................................................................... 23

    1.1. Moving from passive to active targeting ................................................................. 25

    2. Methodology for immunoliposomes development ......................................................... 29

    2.1. Antibody fragments for liposome coupling ......................................................................... 31

    2.2. Coupling methods: Conventional and Post insertion methods ............................................. 33

    2.3. Role of PEG in targeted liposomes ...................................................................................... 35

    3. Mechanism of action ...................................................................................................... 38

    4. Pharmacokinetics (PK) of targeted liposomes................................................................ 40

    5. Clinical trials with targeted liposomes ........................................................................... 45

    5.1. Transferrin-targeted liposomes ............................................................................................ 45

    5.2. HER-2 and EGFR targeted liposomes ................................................................................. 47

    5.3. GAH targeted liposomes ...................................................................................................... 48

    5.4. EphA2 targeted liposomes ................................................................................................... 49

    5.5. Glutathione targeted liposomes ............................................................................................ 50

    6. Future insight for immunoliposomes .............................................................................. 50

    6.1. Patients stratification for immunoliposomes administration ................................................ 50

    6.2. EPR modulation to improve tumor targeting ....................................................................... 51

    6.3. Stimuli-responsive liposomes .............................................................................................. 53

    6.3.1. External stimuli ........................................................................................................................54

    6.3.2. Internal stimuli .........................................................................................................................55

  • CONTENTS

    6.4. Multi-targeting liposomes .................................................................................................... 56

    7. Conclusion ...................................................................................................................... 57

    8. Acknowledgements ........................................................................................................ 59

    9. References ...................................................................................................................... 59

    OBJECTIVES..................................................................................................................... 73

    CHAPTER 2

    A new immune-nanoplatform for promoting adaptive antitumor immune

    response

    Abstract ......................................................................................................................... 81

    Introduction ................................................................................................................... 83

    Materials ........................................................................................................................ 84

    Methods ........................................................................................................................ 85

    1. Preparation of ligands .................................................................................................... .85

    1.1. Anti-PD-L1 mAb ................................................................................................................. 85

    1.2. Anti-PD-L1 Fab’ fragments ................................................................................................ 86

    2. Preparation of immunoliposomes ................................................................................. 86

    3. Coupling methods .......................................................................................................... 87

    3.1. Conventional method ........................................................................................................... 87

    3.2. Post-insertion method .......................................................................................................... 88

    4. Characterization of liposomes ........................................................................................ 89

    5. Stability assay ................................................................................................................. 90

    6. Affinity of ILs for PD-L1 ............................................................................................... 91

    7. In vitro cell studies ......................................................................................................... 91

    7.1. PD-L1 expression ................................................................................................................. 91

    7.2.Cellular interaction of liposomes ......................................................................................... 92

    8. In vivo studies ................................................................................................................. 92

    8.1. Biodistribution study ............................................................................................................ 93

    8.2. Immunological effect ........................................................................................................... 94

    8.3. Therapeutic efficacy ............................................................................................................. 95

    9. Statistical analysis .......................................................................................................... 95

    Results ........................................................................................................................... 96

  • CONTENTS

    1. Enzymatic digestion using pepsin provided Fab’ fragments ......................................... 96

    2. Drug release was not affected by the amount of PEG ................................................... 97

    3. Immunoliposomes bound selectively to PD-L1 ............................................................ 98

    4. In vitro studies ............................................................................................................... 99

    4.1. Baseline PD-L1 expression in inducible by IFN-γ ............................................................... 99

    4.2.ILs showed PD-L1 specificity in B16OVA cells ............................................................... 100

    5. In vivo studies .............................................................................................................. 102

    5.1. Biodistribution patterns evaluated for ILs and non-targeted formulation .......................... 102

    5.2. Fab’ ILs induced a specific immune response ................................................................... 106

    5.3. Fab’ ILs were able to induce tumor shrinkage ................................................................... 107

    Discussion ................................................................................................................... 109

    Acknowledgements ..................................................................................................... 113

    Grant and financial support ......................................................................................... 113

    References ................................................................................................................... 114

    Supplementary material ............................................................................................... 117

    CHAPTER 3

    Doxorubicin immunoliposomes against PD-L1 enhance the immune

    stimulation against melanoma cancer cells

    Abstract ......................................................................................................................... 131

    Introduction .................................................................................................................. 133

    Materials ....................................................................................................................... 134

    Methods ........................................................................................................................ 135

    1. Formulation of Dox liposomes ..................................................................................... 135

    1.1. Anti-PD-L1 Fab’ fragments obtaining .............................................................................. 136

    1.2. Doxorubicin immunoliposomes obtaining ......................................................................... 136

    2. Characterization of liposomes ...................................................................................... 137

    3. Dox release from liposomes ......................................................................................... 138

    4. Cell line ....................................................................................................................... 138

    5. In vitro studies ............................................................................................................. 139

    5.1. Cytotoxicity study .............................................................................................................. 139

    5.2. Cellular interaction of liposomes ....................................................................................... 139

    6. In vivo studies .............................................................................................................. 140

  • CONTENTS

    6.1. Pharmacokinetic evaluation ............................................................................................. 141

    6.2. Activation of the immune system by ILs treatment ......................................................... 142

    6.3. Antitumor effect ............................................................................................................... 144

    7. Statistical analysis ........................................................................................................ 144

    Results .......................................................................................................................... 145

    1. Preparation and characterization of immunoliposomes ............................................... 145

    2. In vitro studies .............................................................................................................. 145

    2.1. Doxorubicin liposomes were stable in serum .................................................................. 145

    2.2. LDF significantly decreased the IC50 ............................................................................... 146

    2.3. PD-L1+ cells showed an increased uptake of targeted liposomes .................................... 147

    3. In vivo studies ............................................................................................................... 149

    3.1. PK profile depends on treatment and dose repetition ...................................................... 149

    3.2. LDF achieved a systemic antitumor immune activation ................................................. 150

    3.3. LDF achieved to control the tumor growth ..................................................................... 152

    Discussion .............................................................................................................. 153

    Acknowledgements ................................................................................................ 158

    Grant and financial support .................................................................................... 158

    References .............................................................................................................. 158

    GENERAL DISCUSSION ............................................................................................... 163

    References ................................................................................................................... 168

    CONCLUSIONS/CONCLUSIONES ............................................................................. 171

    ANNEX I ........................................................................................................................... 177

    ANNEX II .......................................................................................................................... 195

    ANNEX III ........................................................................................................................ 209

  • ABBREVIATIONS

    1

    ABBREVIATIONS

    ABC Accelerated Blood Clearance

    ACK Ammonium Chloride Potassium lysing buffer

    ADAs Anti-drug antibodies

    AUC Area under the curve plasma concentrations

    CH Cholesterol

    CHEMS Cholesteryl hemisuccinate

    DAPI 4′,6-diamidino-2-phenylindole

    DiI 1,1′-Dioctadecyl-3,3,3’,3’-tetramethylindocarbocyanine perchlorate

    DOPE Dioleoylphosphatidylethanolamine

    Dox Doxorubicin

    DPPC Dipalmitoylphosphatidylcholine

    DPPG2 1,2-Dipalmitoyl-sn-glycero-3-phosphatidylglycerol

    DSPC 1,2-Distearoyl-sn-glycero-3-phosphocholine

    DSPE-PEG2000 1,2-Distearoyl-sn-Glycero-3-Phosphoethanolamine-N-

    methoxy(polyethylene glycol)-2000

    EGFR Epidermal Growth Factor Receptor

    EPR Enhanced permeability and retention effect

    Fab´ Monovalent Antibody binding fragment

    F(ab’)2 Bivalent Antibody binding fragment

    FBS Fetal bovine serum

    Fc´ Crystallizable fragment of an antibody

    GrzB Granzyme B

    HER-2 Human Epidermal Growth Factor Receptor -2

    HPLC High-Performance Liquid Chromatography

  • ABBREVIATIONS

    2

    HSPC Hydrogenated Soy L-α-phosphatidylcholine

    Hz-PEG Hydrazine-Polyethylene glycol

    IC Immune Checkpoint

    IC50 Inhibitory concentration 50

    ICD Immunogenic Cell Death

    IL Immunoliposome

    mAb Monoclonal antibody

    Mal-PEG Maleimide-DSPE-PEG2000

    MEA Cysteamine hydrochloride / 2-Mercaptoethylamine HCl

    MIF Mean Intensity Fluorescence

    MWCO Molecular Weight Cut Off

    PD Pharmacodynamics

    PD-1 Programmed Death 1

    PD-L1 Programmed Death Ligand 1

    PDP-PEG Pyridylditiopropionoylamino-Polyethylene glycol

    PEG Polyethylene glycol

    PK Pharmacokinetic

    RES Reticuloendothelial system

    RPMI Roswell Park Memorial Institute medium

    RT Room temperature

    scFv Single chain antibody variable regions

    SD Standard deviation

    SDS Page Sodium dodecyl sulfate-polyacrylamide gel electrophoresis

    SEM Standard Error of the Mean

    SPDP 3-(2-Pyridyldithio) propionic-acid-N-hydroxysuccinimide-ester

  • ABBREVIATIONS

    3

    SRB Sulforhodamine B

    TBS Tris Buffer Saline

    TCEP Tris (2-carboxyethyl) phosphine

    5-FU 5 -Fluorouracil

  • INTRODUCTION

  • INTRODUCTION

    7

    INTRODUCTION

    Cancer is one of the principal causes of death in developed countries 1. Most of the

    anticancer agents used nowadays exert interesting antitumor activity, associated with

    serious side effects that limit, in some cases, the use of these therapies 2,3

    . Chemotherapy is

    characterized by having a large volume of distribution that leads to the dissemination and

    death of both, healthy and malignant cells, without discrimination across them. The

    concept of fighting cancer has changed over the years in order to reduce toxicity,

    improving the life quality of the patients treated with chemotherapy. In this way, the

    encapsulation of these molecules in different types of nanocarriers, in particular liposomes,

    has provided more specific and safe treatments, some of which have achieved clinical

    application 4,5

    . Liposomal formulations are capable of increasing drug efficiency while

    reducing toxicity 6. These nanocarriers, often composed of cholesterol (CH) and

    phospholipids, are biocompatible and low immunogenic vehicles that can carry or

    transport, by encapsulation, a large number of molecules with different physicochemical

    properties 4,6

    . The main advantage of liposomes is their capability to be accumulated in the

    tumor area, enhancing the therapeutic drug activity. This is a consequence of changes in

    the pharmacokinetics (PK) properties of the encapsulated agent, in particular a reduction in

    the distribution. In addition, the modification of current liposomes by the inclusion of

    certain polymers, such as polyethyleneglycol (PEG) covering their surface, provides an

    increment in their circulation half-life in comparison to early liposomes, which were

    rapidly recognized and removed by the reticuloendothelial system (RES) 4,7,8

    .

    Pegylated liposomes have some favourable characteristics for cancer therapy, such as low

    toxicity, improved PK, long-term circulation in blood and tumor accumulation, this last

    one promoted by the Enhanced Permeability and Retention effect (EPR) (Figure 1) 4,6,9

    .

    The EPR effect is based on the presence of a high permeability on the vascular

  • INTRODUCTION

    8

    endothelium in tumor due to the rapid growth of vessels, which results in the formation of

    fenestrated and leaky blood vessels. This characteristic enables the extravasation of

    molecules with a size up to 300 nm to the tumor area, which are accumulated there due to

    the lack of lymphatics 10–12

    .

    Figure 1. Schematic representation of the EPR effect: A) Healthy tissue; B) Tumor tissue.

    However, despite the tumor accumulation, generally, liposomal formulations present high

    stability, limiting the intracellular drug bioavailability, and thereby the antitumor effect is

    suboptimal. This fact has led to modify these liposomes by the inclusion of different types

    of lipids in their composition, which allow the control of the drug release. Thus, when

    certain stimulus is applied (ex. temperature, light) or is present in the tumor (pH change,

    enzymes), these lipids change their conformation and destabilize the liposome membrane,

    driving to the rapid release of the cargo 4,13–16

    . However, despite these advantages of

    improving the rapid release at the tumor site, the lack of selectivity and specificity of these

  • INTRODUCTION

    9

    stimulus-sensitive formulations may lead to toxicities in tumor surrounding healthy tissues

    by the non-internalized drug, which is cleared by systemic circulation 6,17

    .

    In this regard, liposomes can be functionalized with different molecules (antibodies,

    peptides, proteins or carbohydrates) attached to their surface to obtain targeted liposomes,

    triggering a specific tumor cell recognition and interaction. However, to achieve a

    therapeutic benefit, the target of the ligand coupled to liposomes must be overexpressed,

    upregulated or be exclusive of tumor cells in response to a high metabolic demand or

    mutations 4. Thus, liposomes targeted with certain proteins, aptamers or carbohydrates

    allow tumor-specific delivery to the target sites, bypassing normal healthy cells 6,9

    .

    Figure 2. Schematic representation of the different fragments of a monoclonal antibody: A) Whole

    mAb; B) F(ab’)2 fragment; C) Fab’ fragment; D) ScFv fragment.

    This procedure triggers an increment on the therapeutic index of the encapsulated agent

    6,17, being accumulated at the tumor site (based on the EPR effect) and facilitating the

    specific uptake of targeted liposomes by malignant cells 6. Among the different ligands,

    monoclonal antibodies (mAbs) and their monovalent variable fragments are the most

    widely targeting moieties (Figure 2) used to develop targeted liposomes, known as

    immunoliposomes (ILs) (Figure 3) 4,6,9,17

    .

  • INTRODUCTION

    10

    Figure 3. Evolution of liposomes: A) Plain liposome; B) Pegylated long-circulating liposome; C) ILs.

    Although ILs showed to be more advantageous formulation than non-targeted liposomes,

    few have been commercialized. Currently, those conjugated with HER-2, GAH and

    Epidermal Growth Factor (EGF) are involved in several clinical trials with promising

    results 4,17

    .

    In this strategy, the liposome acts as a drug carrier and the ligand, such as the antibody, as

    the driver to anchor the formulation to the target 17

    . Nevertheless, this work proposes a step

    further in ILs antitumor fight: the use of an antibody moiety not only to target but also to

    exert a specific effect. In that sense, Immunotherapy is emerging as a promising approach

    against many types of cancer 1, relying on the capacity of the immune system to eliminate

    cancer cells 18

    (Figure 5).

    The immune system is able to identify specific tumor antigens from highly immunogenic

    cancer cells, and kill them at an incipient status by the immunosurveillance process, which

    shapes the cycle of immunity described by Chen & Mellman (2013) 19–21

    . However, tumors

    composed of weakly immunogenic malignant cells are characterized by a high frequency

    of genetic and epigenetic abnormalities, which lead to the establishment of a process

    referred to as “cancer immunoediting” 19,21

    , which enables tumor escape from the immune

    system. In this case, tumors provide mechanisms to transform effector T cells in non-

    effective cells either by inactivation or by hyper-activation, leading to their dysfunction. In

    this scenario, several factors in the tumor microenvironment can negatively modulate the

    activated antitumor T cells, contributing to the immune escape of malignant cells 22

    .

  • INTRODUCTION

    11

    Immune checkpoint (IC) molecules such as Programmed Death-1 (PD-1) and its ligand,

    Programmed Death-Ligand 1 (PD-L1) 23

    are involved in the crosstalk of immune cells and

    tumor. PD-L1 is a molecule commonly overexpressed in tumor cells that binds to PD-1, a

    receptor present in activated T cells. PD-L1/PD-1 binding downregulates T cell activity,

    leading to T cell exhaustion or dysfunction 18,24,25

    . According to this feature, several

    monoclonal antibodies have been approved to block these ICs and can be conjugated into

    ILs in order to control the immunosuppressive mechanisms 19,26

    (Figure 4).

    Figure 4. Representation of the interaction mechanism between PD-1/PD-L1: A) Downregulation of T

    cell activity; B) Reactivation of the immune system.

    Thus, Pembrolizumab and Nivolumab, which recognize and bind PD-1, together with

    Atezolizumab, an anti-PD-L1 mAb recently approved by the FDA for the treatment of

    mesothelial tumors, have demonstrated impressive clinical outcomes 23,25,27–29

    . However,

  • INTRODUCTION

    12

    their use in monotherapy has led to a low amount of patients who experience therapeutic

    benefits, supporting the necessity of introducing combinatorial approaches 23,30

    .

    In this way, some chemotherapeutic drugs, like Doxorubicin (Dox), have the ability to

    induce immunogenic cell death (ICD) producing apoptosis and inflammation 31,32

    , which

    contributes to stimulate the immune system against tumor cell-death antigens, leading to

    the activation of T cells 32–34

    (Figure 5). Thus, the combination of ICD compounds with

    immunotherapy, like IC inhibitors, is coined Chemoimmunotherapy. This is a new

    emerging rational therapeutic approach 35

    that has not been assayed in depth yet. However,

    this association may provide a comprehensive understanding of the underlying antitumor

    mechanisms.

    Figure 5. Schematic representation of the immune response after chemoimmunotherapy treatment.

    (CRT: Calreticulin; HMGB1: High-mobility group box 1 protein; ATP: Adenosine triphosphate; ICD:

    Immunogenic Cell Death; IFN-γ: Interferon-gamma; IL-2: Interleukin-2; TNF: Tumor Necrosis

    Factor)

    According to this, the proposal of this work has been the combination of immunotherapy

    and chemotherapy, applying the advantages of nanotechnology. Thus, we hypothesize that

    liposomes encapsulating Dox and coupled to anti-PD-L1 at their surface would induce

    tumor regression by a dual mechanism, specifically killing malignant cells and stimulating

  • INTRODUCTION

    13

    the immune system by the blockage of PD-L1, that would reduce the immunosuppressive

    environment 9. In our knowledge, this is a new concept for ILs that represents the main

    contribution of this project.

    To achieve the present aim, several steps represented in the different chapters have been

    addressed. Firstly, a detailed review about the state of the art of ILs, as well as a summary

    about the main key points that have to be controlled to develop adequate ILs, is reported in

    Chapter 1.

    Chapter 2 is focused on the development of different immunoliposomes coupled with the

    anti-PD-L1 mAb and its Fab’ fragment, using different coupling methods and PEG

    percentages, to be later assayed in in vitro / in vivo studies.

    Here, factors such as drug encapsulation and ligand density have been optimized, making

    an important effort to achieve a formulation able to deal with adequate characteristics to

    reach the tumor area and produce efficient intracellular drug delivery and immune activity.

    Finally, in Chapter 3, the formulation selected in the previous work has been combined

    with Dox as a proof of concept, to evaluate its in vitro / in vivo activity. This immune-

    nanoplatform encapsulating Dox and targeted against PD-L1 showed a dual mechanism

    characterized by an immune stimulation and a cytotoxic activity, both contributing to

    reduce and eliminate cancer cells.

    REFERENCES

    1. Quetglas JI, Hervas-Stubbs S, Smerdou C. The immunological profile of tumor-bearing

    animals determines the outcome of cancer immunotherapy. Oncoimmunology.

    2013;2(6):e24499. doi:10.4161/onci.24499.

    2. Remesh A. Toxicities of anticancer drugs and its management. Int J Basic Clin Pharmacol.

    2012;1(1):2. doi:10.5455/2319-2003.ijbcp000812.

    3. Chatelut E, Delord J-P, Canal P. Toxicity patterns of cytotoxic drugs. Invest New Drugs.

    2003;21(2):141-148. http://www.ncbi.nlm.nih.gov/pubmed/12889735.

  • INTRODUCTION

    14

    4. Merino M, Zalba S, Garrido MJ. Immunoliposomes in clinical oncology: State of the art and

    future perspectives. J Control Release. 2018;275:162-176.

    doi:10.1016/j.jconrel.2018.02.015.

    5. Eloy JO, Claro de Souza M, Petrilli R, Barcellos JPA, Lee RJ, Marchetti JM. Liposomes as

    carriers of hydrophilic small molecule drugs: strategies to enhance encapsulation and

    delivery. Colloids Surf B Biointerfaces. 2014;123:345-363.

    doi:10.1016/j.colsurfb.2014.09.029.

    6. Tila D, Ghasemi S, Yazdani-Arazi SN, Ghanbarzadeh S. Functional liposomes in the

    cancer-targeted drug delivery. J Biomater Appl. 2015;30(1):3-16.

    doi:10.1177/0885328215578111.

    7. Owens DE, Peppas NA. Opsonization, biodistribution, and pharmacokinetics of polymeric

    nanoparticles. Int J Pharm. 2006;307(1):93-102. doi:10.1016/j.ijpharm.2005.10.010.

    8. Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving

    nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev. 2016;99:28-51.

    doi:10.1016/j.addr.2015.09.012.

    9. Eloy JO, Petrilli R, Trevizan LNF, Chorilli M. Immunoliposomes: A review on

    functionalization strategies and targets for drug delivery. Colloids Surf B Biointerfaces.

    2017;159:454-467. doi:10.1016/j.colsurfb.2017.07.085.

    10. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors

    as well as issues related to its heterogeneity. Adv Drug Deliv Rev. 2015;91:3-6.

    doi:10.1016/j.addr.2015.01.002.

    11. Iyer AK, Khaled G, Fang J, Maeda H. Exploiting the enhanced permeability and retention

    effect for tumor targeting. Drug Discov Today. 2006;11(17-18):812-818.

    doi:10.1016/j.drudis.2006.07.005.

    12. Maeda H. The enhanced permeability and retention (EPR) effect in tumor vasculature: The

    key role of tumor-selective macromolecular drug targeting. Adv Enzyme Regul.

    2001;41:189-207. doi:10.1016/S0065-2571(00)00013-3.

    13. Haeri A, Zalba S, ten Hagen TLM, Dadashzadeh S, Koning GA. EGFR targeted

    thermosensitive liposomes: A novel multifunctional platform for simultaneous tumor

    targeted and stimulus responsive drug delivery. Colloids Surfaces B Biointerfaces.

    2016;146:657-669. doi:10.1016/j.colsurfb.2016.06.012.

    14. Kono K, Takashima M, Yuba E, et al. Multifunctional liposomes having target specificity,

    temperature-triggered release, and near-infrared fluorescence imaging for tumor-specific

    chemotherapy. J Control Release. 2015;216:69-77. doi:10.1016/j.jconrel.2015.08.005.

    15. Guo P, You J-O, Yang J, Jia D, Moses MA, Auguste DT. Inhibiting metastatic breast cancer

    cell migration via the synergy of targeted, pH-triggered siRNA delivery and chemokine axis

  • INTRODUCTION

    15

    blockade. Mol Pharm. 2014;11(3):755-765. doi:10.1021/mp4004699.

    16. Li Q, Tang Q, Zhang P, et al. Human epidermal growth factor receptor-2 antibodies

    enhance the specificity and anticancer activity of light-sensitive doxorubicin-labeled

    liposomes. Biomaterials. 2015;57:1-11. doi:10.1016/j.biomaterials.2015.04.009.

    17. Sapra P, Shor B. Monoclonal antibody-based therapies in cancer: advances and challenges.

    Pharmacol Ther. 2013;138(3):452-469. doi:10.1016/j.pharmthera.2013.03.004.

    18. Merelli B, Massi D, Cattaneo L, Mandalà M. Targeting the PD1/PD-L1 axis in melanoma:

    biological rationale, clinical challenges and opportunities. Crit Rev Oncol Hematol.

    2014;89(1):140-165. doi:10.1016/j.critrevonc.2013.08.002.

    19. Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new

    immunomodulatory targets. Nat Rev Drug Discov. 2015;14(8):561-584.

    doi:10.1038/nrd4591.

    20. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity.

    2013;39(1):1-10. doi:10.1016/j.immuni.2013.07.012.

    21. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and

    immunoediting. Immunity. 2004;21(2):137-148. doi:10.1016/j.immuni.2004.07.017.

    22. Chang C-H, Qiu J, O’Sullivan D, et al. Metabolic Competition in the Tumor

    Microenvironment Is a Driver of Cancer Progression. Cell. 2015;162(6):1229-1241.

    doi:10.1016/j.cell.2015.08.016.

    23. Contreras-Sandoval AM, Merino M, Vasquez M, Trocóniz IF, Berraondo P, Garrido MJ.

    Correlation between anti-PD-L1 tumor concentrations and tumor-specific and nonspecific

    biomarkers in a melanoma mouse model. Oncotarget. 2016;7(47):76891-76901.

    doi:10.18632/oncotarget.12727.

    24. Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to

    activate anti-tumor immunity. Curr Opin Immunol. 2012;24(2):207-212.

    doi:10.1016/j.coi.2011.12.009.

    25. Robert C, Soria J-C, Eggermont AMM. Drug of the year: programmed death-1

    receptor/programmed death-1 ligand-1 receptor monoclonal antibodies. Eur J Cancer.

    2013;49(14):2968-2971. doi:10.1016/j.ejca.2013.07.001.

    26. Blank CU. The perspective of immunotherapy: new molecules and new mechanisms of

    action in immune modulation. Curr Opin Oncol. 2014;26(2):204-214.

    doi:10.1097/CCO.0000000000000054.

    27. Borch TH, Donia M, Andersen MH, Svane IM. Reorienting the immune system in the

    treatment of cancer by using anti-PD-1 and anti-PD-L1 antibodies. Drug Discov Today.

    2015;20(9):1127-1134. doi:10.1016/j.drudis.2015.07.003.

    28. Philips GK, Atkins M. Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies. Int

  • INTRODUCTION

    16

    Immunol. 2015;27(1):39-46. doi:10.1093/intimm/dxu095.

    29. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature.

    2011;480(7378):480-489. doi:10.1038/nature10673.

    30. Shi T, Ma Y, Yu L, et al. Cancer Immunotherapy: A Focus on the Regulation of Immune

    Checkpoints. Int J Mol Sci. 2018;19(5). doi:10.3390/ijms19051389.

    31. Casares N, Pequignot MO, Tesniere A, et al. Caspase-dependent immunogenicity of

    doxorubicin-induced tumor cell death. J Exp Med. 2005;202(12):1691-1701.

    doi:10.1084/jem.20050915.

    32. Tesniere A, Apetoh L, Ghiringhelli F, et al. Immunogenic cancer cell death: a key-lock

    paradigm. Curr Opin Immunol. 2008;20(5):504-511. doi:10.1016/j.coi.2008.05.007.

    33. Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy.

    Annu Rev Immunol. 2013;31:51-72. doi:10.1146/annurev-immunol-032712-100008.

    34. Locher C, Conforti R, Aymeric L, et al. Desirable cell death during anticancer

    chemotherapy. Ann N Y Acad Sci. 2010;1209:99-108. doi:10.1111/j.1749-

    6632.2010.05763.x.

    35. Emens LA. Chemoimmunotherapy. Cancer J. 16(4):295-303.

    doi:10.1097/PPO.0b013e3181eb5066.

  • CHAPTER 1

    IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE

    OF THE ART AND FUTURE PERSPECTIVES

  • CHAPTER 1

    IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART

    AND FUTURE PERSPECTIVES

    María Merino1, Sara Zalba

    1, María J. Garrido

    1*

    1Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy and Nutrition,

    University of Navarra, Pamplona, Spain

    Published in Journal of Controlled Release

    Keywords: Liposome, Monoclonal Antibody, pharmacokinetic characteristics, Clinical

    applications.

    *Corresponding author: Address: Department of Pharmacy and Pharmaceutical Technology,

    University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain. Tel.: +34 948425600: 6529. E-mail

    address: [email protected] (María J. Garrido)

  • CHAPTER 1

    21

    ABSTRACT

    Liposomal formulations entrapping a vast number of molecules have improved cancer

    therapies overcoming certain pharmacokinetic (PK) and pharmacodynamic limitations,

    many of which are associated with tumor characteristics. In this context,

    immunoliposomes represent a new strategy that has been widely investigated in

    preclinical cancer models with promising results, although few have reached the stage

    of clinical trials. This contrasts with the emerging clinical application of monoclonal

    antibodies (mAbs). This formulation allows the conjugation of different mAbs or

    antibody derivatives, such as monovalent variable fragments Fab’, to the polymers

    covering the surface of liposomes. The combination of this targeting strategy together

    with drug encapsulation in a single formulation may contribute to enhance the efficacy

    of these associated agents, reducing their toxicities.

    In this paper, we will consider how factors such as particle size, lipid composition, and

    charge, lipid-polymer conjugation, method of production and type of ligand for

    liposome coupling influence the efficacy of these formulations. Furthermore, the high

    inter-individual variability in the tumor microenvironment, as well as the poor

    experimental designs for the PK characterization of liposomes, makes the establishment

    of the relationship between plasma or tumor concentrations and efficacy difficult. Thus,

    adequate dosing regimens and patient stratification regarding the target expression may

    contribute to enhance the possibility of incorporating these immunoliposomes into the

    therapeutic arsenal for cancer treatments. All these issues will be briefly dealt with here,

    together with a section showing the state of the art of those targeted liposomes that are

    coming up for testing in clinical trials. Finally, some insights into future developments

    such as the combination of specificity and controlled release, based on the application of

    different stimuli, for the manipulation of stability and cargo release, will be offered.

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    22

    This has been included in order to highlight the new opportunities for targeted

    liposomes, including immunoliposomes.

  • CHAPTER 1

    23

    1. INTRODUCTION

    Over the years, chemotherapy based on conventional anticancer drugs has demonstrated

    good clinical efficacy. However, these molecules present serious side effects associated

    with their wide and non-specific body distribution, leading in general, to a dose-

    limitation or discontinuation of treatment. Thus, in order to reduce chemotherapeutic

    toxicity, some of these drugs have been encapsulated in nanoparticles such as liposomes

    1.

    Liposomes are biocompatible and lowly immunogenic hollow vesicles usually made up

    of phospholipids and cholesterol (CH). They are able to encapsulate a wide range of

    drugs 2,3

    that provide important advantages, in particular, those related to

    pharmacokinetics, such as the reduction of the volume of distribution and the extension

    of blood circulation time. All of these contribute to the enhancement of the therapeutic

    index, providing a higher accumulation in the tumor and protecting healthy tissues from

    side effects 1,2

    .

    However, conventional liposomes, rapidly recognized by opsonins and removed by the

    reticuloendothelial system (RES), were modified by the addition of gangliosides such as

    GM1 4 or polymers as is the case of polyethylene glycol (PEG). This change entails the

    presence of a hydrophilic layer surrounding the liposome, delaying opsonization and,

    thereby, RES removal (see Figure 1). This leads to an increase of the circulation half-

    life of the formulation from hours to days, forming the so-called long-circulating

    liposomes 5,6

    .

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    24

    Figure 1. Schematic representation of the opsonization process. Opsonized liposomes are able to

    adhere to phagocytes through opsonin receptors, as mannose and scavenger receptors 7, and are

    removed by the RES in the case of: A) conventional or non-pegylated liposomes; B) pegylated

    liposomes, which are described in particular detail in the schema represented in panel C.

    Since these modified liposomes have demonstrated to equal or even improve the

    antitumor effect compared to the free drug in preclinical and clinical studies 2, several

    liposomal formulations have been approved for the treatment of different diseases.

    Table 1 lists some of the current liposomal formulations in clinical use or involved in

    clinical trials.

  • CHAPTER 1

    25

    Table 1. The most popular approved liposomal formulations in clinical oncology.

    Active drug Product name Indication

    Annamycin Annamycin liposomes Doxorubicin-resistant tumors

    Cytarabine Depocyt Cancer therapy

    Daunorubicin

    Daunoxome

    AIDS-related Kaposi’s sarcoma, breast and lung cancer

    Daunorubicin/ Cytarabine Vyxeos Acute myeloid leukemia

    Doxorubicin (non-PEG liposomes)

    Myocet

    Combinatorial therapy for recurrent breast cancer

    Doxorubicin (PEG- liposomes) Doxil/ Caelyx

    Refractory Kaposi’s sarcoma, refractory ovarian cancer, recurrent breast cancer,

    multiple myeloma

    Doxorubicin Evacet Metastatic breast cancer

    Irinotecan Onivyde® Metastatic pancreatic cancer

    Lurtotecan NX211 Ovarian cancer

    Plasmid encoding HLA-B7 and β2 microglobulin

    Allovectin-7 * Metastatic melanoma

    Platinum derivative (NDDP) 8 Platar Solid tumors

    Tretinoin AtragenTM

    Kaposi’s sarcoma , acute Pro-myelocytic leukemia, non-

    Hodgkin’s lymphoma, renal cell carcinoma

    Vincristine Marqibo® Acute lymphoblastic leukemia

    Vincristine Onco TCS Non-Hodgkin’s lymphoma

    Vincristine VincaXome Solid tumors

    * Allovectin-7 is an immunotherapeutic agent; NDDP: cis-bis-neodecanoato (trans- R, R-1, 2-diaminocyclohexane)-

    platinum II.

    1.1. Moving from passive to active targeting

    Cancer cells, characterized by rapid replication, have a high demand for nutrients and

    oxygen for tumor growth. In this process, there is acidification and hypoxia in the tumor

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    26

    microenvironment. These stimuli promote an increase in tumor vascularization, known

    as angiogenesis or neovascularization. However, this neovascularization results in

    immature tumor vessels that differ from healthy vessels 9,10

    with regard to their

    permeability and organization. Tumor vessels lack the tight junctions between

    endothelial cells, giving rise to fenestrated and leaky blood vessels, irregular in shape,

    with no smooth-muscle layer 9–11

    . These particular characteristics allow the

    extravasation of particles with a size up to 300 nm from blood to the tumor area. This,

    together with the absence of proper lymphatic drainage, leads to a higher accumulation

    of long-circulating nanoparticles in the tumor. This represents the basis of the Enhanced

    Permeability and Retention (EPR) effect, responsible for the Passive accumulation of

    liposomes and other nanoparticles in tumor tissue 9,10,12

    (more detailed info in Section

    6.2).

    However, there is not always a correlation between the presence of nanoparticles in the

    tumor microenvironment and an increase in intracellular drug bioavailability 13,14

    . This

    may be due to two different situations. The first can be related to the high stability of

    liposomes, which leads to poor and slow drug release; whereas the second might be

    associated with the low cellular uptake efficiency of liposomes. To overcome this last

    obstacle, “Active Targeting” o ligand-based targeting 3,14–16

    has emerged as one of the

    most promising strategies to improve specific drug internalization 17

    . In this approach,

    targeted liposomes are able to directly bind to cancer cells. For this, the surface of the

    liposomal formulations has to be decorated with different types of ligands, which

    specifically recognize and bind those molecules expressed or over-expressed on the

    surface of cancer cells 18

    . Thus, some of these molecules, such as growth factor

    receptors, are upregulated in response to an increased metabolic demand and are

    associated with a poor prognosis in cancer. In addition, there are also certain epitopes,

  • CHAPTER 1

    27

    antigens, or receptors on the membrane of tumor cells which represent attractive targets

    for the functionalization of liposomes 4,11

    . Epidermal Growth Factor Receptor (EGFR),

    Folate Receptor (FR) or Transferrin Receptors (TfR), among others, represent the most

    commonly used receptors for liposome targeting. In this way, Epidermal Growth Factor

    (EGF), Folate and Transferrin are the corresponding ligands for decorating these

    liposomes 7.

    However, due to the high mutational rate, cancer cells also express exclusive membrane

    markers suitable for targeting, which improve the specificity of these targeted

    liposomes. As a result, the establishment of a parallel process involving the

    identification of these molecules for developing specific agents or ligands to block or

    inhibit their activities is of particular interest 19

    . Thus, the impact of DNA-techniques

    for engineering more specific molecules has been particularly marked in the

    development of monoclonal antibodies (mAbs) 20

    . At that point, the coupling of mAbs

    to the surface of liposomes represents the basis of “Immunoliposomes” 21

    , a particular

    type of targeted liposomes.

    It is worth pointing out that the relevance of mAbs in cancer treatment is increasing

    exponentially, despite their side effects, in particular, immunogenic reactions 22,23

    .

    Combinatorial therapies including mAbs and other agents, such as cytotoxic molecules,

    are very often applied in patients. This is why immunoliposomes may represent an

    interesting strategy, although currently, they are still in preclinical or initial clinical

    phases. Table 2 lists the most common mAbs or antibody fragments conjugated to

    different doxorubicin liposomes, reported in the literature 11

    .

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    28

    Table 2. Summary of the main immunoliposomes reported in the literature for preclinical studies.

    Targeting receptors are alphabetically ordered.

    Receptor Ligand Drug Type of cancer Tumor

    accumulation * Coupling efficiency

    Ref.

    CD19

    mAb Doxorubicin B-lineage

    malignancies (B-cell leukemias / lymphomas and

    Multiple Myeloma)

    N.A. 110 µg mAb/µmol

    PL 24

    mAb Fab’

    Doxorubicin / Vincristine

    3 50-70 % Fab’

    80-90 % mAb

    25

    mAb Doxorubucin /

    Vincristine N.A.

    60 µg mAb/µmol PL

    26

    CD20 mAb Doxorubicin B-cell Lymphoma ND 54 µg mAb/µmol

    PL 24,26

    CD74 Fab’ Doxorubicin B-lymphoma 6 (24h later) 60 Fab’/LP 27

    EGFR Fab’ Doxorubicin

    Epithelial tumors 6 (24h later) 30 µg Fab’/µmol

    LP 28

    GAH F(ab’)2 Doxorubicin Advanced gastric

    cancer N.A.

    0.02 µg F(ab)2/µg lipids

    29

    GD2

    mAb Fab’

    Doxorubicin

    Neuroblastoma 10

    0.33-0.53 nmol mAb/μmol PL

    0.54-0.91 nmol Fab’/µmolPL

    30,31

    mAb c-myb Antisense

    ODN 60-80 µg

    mAb/µmol PL 32

    HER-2 Fab’ scFV

    Doxorubicin Breast cancer N.A. 30-50 Fab’/IL

    20 scFv/IL 33,34

    VEGFR Fab’ Doxorubicin Tumor-associated endothelial cells

    10 300 Fab’/LP 35,36

    2C5 (ANAs)

    mAb Doxorubicin Cancer cell

    specific nucleosomes

    2 (24h later) 80-100 mAb/LP

    37,38

    5D4 mAb Doxorubicin Prostate cancer N.A. 100 mAb/LP 39

    PL: phospholipid; IL: immunoliposome; LP: liposome; EGFR: Epidermal Growth Factor Receptor; GAH:

    Genetically Altered Hybridoma; GD2: Disialoganglioside 2; HER2: Human Epidermal Growth Factor Receptor 2;

    VEGFR: Vascular Endothelial Growth Factor Receptor; ANAs: Antinuclear Autoantibodies; Fab’: Antigen Binding

    Fragment; scFv: single chain Fv; ODN: Oligodeoxynucleotides; N.A.: non-available.

    * fold higher compared with non-targeted liposomes.

  • CHAPTER 1

    29

    Therefore, targeted liposomes have shifted the concept of specificity and selectivity for

    tumor drug delivery, providing higher tumor recognition and subsequent cell uptake

    compared to non-targeted formulations, that may lead to high drug tumor accumulation, as

    is reported in Table 2 18.

    2. METHODOLOGY FOR IMMUNOLIPOSOMES DEVELOPMENT

    Over recent decades, the methods developed for targeted liposome formulation, including

    immunoliposomes, have been changing in order to achieve higher coupling efficiency for

    the different ligands, more stable formulations and methods which may be easily scalable.

    Initially, ligands were directly attached to the headgroups of lipids for conventional

    liposomes. However, the pegylation process changed the concept of ligand coupling.

    Although one of the limitations was low ligand availability due to the ligand being hidden

    in the polymeric cover, currently ligand molecules are exposed to the external surface of

    targeted liposomes, avoiding sterical hindrance and guaranteeing recognition of the target

    1,11,19,25,26. For this reason, ligands are currently attached to the end of PEG chains to

    minimize a possible poor binding capacity to the target. Moreover, the combination of

    PEG with different molecular weights has also been reported by some authors to obtain

    immunoliposomes with a higher targeting efficiency 40–42

    . Thus, the use of one PEG

    derivative that is longer than the other, with only one being functionalized has been

    implemented.

    Therefore, the main strategy that has been adopted is the use of an end-functionalized

    pegylated lipid. This lipid derivative is able to form a covalent bond with the ligands,

    which normally requires previous activation or modification 23,24

    . In general, targeted

    liposomes are formulated with 1% of end-functionalized pegylated lipid. However, the

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    30

    length and amount of PEG play a significant role regarding ligand-target binding ability.

    This issue needs further comment and will be dealt with fully in section 2.3.

    Table 3. Brief summary of the main linkers used for mAb ligand coupling during immunoliposomes

    development.

    PEG-derivatives Strategy Coupling efficiency

    Ligand Ref.

    Hz-DSPE-PEG Oxidation mAb 22-30 µg mAb/µmol PL N.A. 47

    PDP-DSPE-PEG Reduction of PDP groups

    conjugated with mAb-Mal micelles

    93-96 µg mAb/µmol PL N.A. 47

    Mal-DSPE-PEG Thiolation mAb 5-25 µg protein/µmol

    lipid Cetuximab

    or EGF 48,49

    Mal-PEG-CH Thiolation mAb 25-35 µg mAb/µmol

    lipid Cetuximab

    45

    Cyanur-DSPE-PEG PEG nucleophilic substitution 30-35 µg protein/µmol

    lipid

    Anti-E-selectin

    mAb

    50

    Folate-PEG-CHEMS Reaction of

    FBP-mAb with folate-LP 25 µg FBP-C225/µmol

    lipid Cetuximab

    (C225) 51

    PL: phospholipid; FBP: folate binding protein; EGF: epidermal growth factor; LP: liposomes; N.A.: non-available.

    In line with this, ligands such as mAbs have been coupled to immunoliposomes using

    different PEG derivative linkers, as shown in Table 3. For instance, Hydrazide (Hz)-PEG,

    pyridylditiopropionoylamino (PDP)-PEG, maleimide (Mal)-PEG and cyanur-PEG are the

    most common lipids associated with PEG reported in literature. Additionally, folate-PEG-

    CHEMS 43

    , Cholesterol-anchored PEG 44

    , or even the maleimide-PEG-cholesterol (mal-

    PEG-CH) are also used, although they are less relevant 45

    . Moreover, in recent years, it has

    been reported that CH anchors are less stable when they reach the blood stream. This is

    due to their hydrophobicity, which may modify the solubility of the ligands anchored to

    CH, affecting their biodistribution, pharmacokinetics and efficacy. Therefore, the delivery

  • CHAPTER 1

    31

    mechanism of these CH-anchored conjugates may alter their interaction with proteins,

    cells, receptors and membranes 46

    .

    Depending on the linker, covalent and non-covalent binding can be achieved. Thus, PDP-

    PEG, Mal-PEG and Hz-PEG are the most common PEG derivatives for covalent

    conjugation, whereas, Cyanur-DSPE-PEG, Folate-PEG-CHEMS and Biotine-PEG are

    those most frequently used for non-covalent coupling (Table 3).

    Note that for immunoliposomes, the coupling efficiency may be affected by the particular

    method applied for mAb activation, because depending on the location of these activated

    groups, this process can also influence targeting efficiency. Thus, although these activated

    groups are arbitrarily distributed throughout the whole ligand structure, sometimes a

    random orientation is given during liposome conjugation 4,27

    , hindering receptor

    attachment and hence, reducing specificity and targeting efficacy. However, to overcome

    the random orientation of the mAb for the coupling, monovalent variable fragment of

    mAbs, Fab’, can be used.

    On the other hand, there are methodologies providing ligand conjugation without any

    previous modification. These can achieve a higher ligand-receptor binding efficiency, as

    occurs with the cyanuric chloride-DSPE-PEG linker 50

    .

    2.1. Antibody fragments for liposome coupling

    Despite the limitations for the conjugation of the whole mAb to immunoliposomes, this

    strategy is still a very promising therapeutic approach 48

    , even when the presence of both

    the Fc fragment and the required activation of the mAb may decrease residence time in

    blood and formulation specificity, respectively (see in section 4). In order to improve these

    characteristics, Fab’ fragments seemed to be one of the main solutions.

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    32

    Figure 2 shows the process involved in obtaining Fab’. This starts with the enzymatic

    digestion of the mAb to collect the F(ab’)2 fragments, which are then reduced using agents

    such as β-mercaptoethanol or SDS, resulting in the monovalent Fab’ fragment. In this last

    step of the process, F(ab’)2 fragments are also treated with β-mercaptoethylamine

    hydrochloride (MEA), to specifically reduce the sulfhydryl groups between the light chains

    of these bivalent fragments, allowing the exposure of -SH radicals for a correct coupling

    process, and keeping receptor recognition available 24,28

    .

    In our experience, mAb fragmentation using pepsin may be associated with some technical

    problems. This enzyme is difficult to remove during fragment collection and interferes in

    protein quantification and fragment identification in western-blot analyses. At this point,

    the use of immobilized pepsin is highly recommended, ensuring the absence of this protein

    in the samples collected.

    (MEA: 2-Mercaptoethylamine•HCl).

    Figure 2. Schematic representation of two enzymatic digestion processes for obtaining different mAb

    fragments: (A) pepsin and (B) papain.

  • CHAPTER 1

    33

    Smaller mAb fragments such scFv, Single domain antibodies (sdAbs), nanobodies or a

    Phage Display Library have also been used for this targeting approach 52–54

    . The lower

    molecular weight of these ligands allows the number of molecules conjugated per liposome

    to be increased in order to compensate for the monovalency of these molecules 55,56

    .

    Nevertheless, it has been widely reported that only 10-20 molecules of ligand per liposome

    seem sufficient to achieve an efficient targeting effect 55

    .

    2.2. Coupling methods: Conventional and Post-insertion methods

    Immunoliposomes can be formulated following the two main methods reported in the

    literature, as is shown in figure 3:

    Conventional method: This includes a DSPE-PEG derivative in the lipid composition

    of liposomes, providing that approximately 50% of this end-functionalized PEG is

    orientated to the inner space of the formulation; ligands such as mAb or Fab’ need to

    be activated for their attachment to the previously formulated liposomes.

    Post-insertion method: This consists of two steps: first, an end-functionalized DSPE-

    PEG derivative forms micelles, which are coupled to the ligand 19,30

    ; second, targeted

    micelles are incorporated into previously developed liposomes by a single incubation

    29.

    In general, the main advantage of the post-insertion strategy is the flexibility for the ligand

    to be coupled to any type of liposomes, encapsulating different therapeutic agents in a

    single step.

    However, for both methods, the stability of the end-derivative lipid must be considered

    during these coupling procedures. Thus, although 50-63% of active maleimide groups

    remain on the surface for the conventional method, their activity decreases to 32% during

    the purification and coupling processes. In contrast, for the post-insertion method, this

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    34

    effect is minimized, with 76% of maleimide groups showing activity for the coupling

    reaction 57

    . However, a small amount (2-5%) of the encapsulated drug during these

    processes might be lost 58

    .

    Figure 3. Schematic representation of the two main methods described for the development of

    immunoliposomes coupled to mAb or Fab’-fragment. A) Conventional method; B) Post-insertion

    method.

  • CHAPTER 1

    35

    2.3. Role of PEG in targeted liposomes

    At the early stages of liposome development, the erythrocyte glycocalyx was mimicked to

    obtain stealth liposomes by using polysaccharides, gangliosides and hydrated

    phosphatidylinositol 59,60

    . However, in order to obtain a feasible procedure for large-scale

    manufacture, several polymers were tested to prolong the circulation time of nanoparticles

    in blood 61

    . For this reason, the PEG polymer was selected and, currently, it is the most

    widely used in pharmaceutical applications due to its biocompatibility and stealth

    properties. The molar mass of PEG is variable depending on the type of conjugation, 20-50

    kDa for small molecules, and 1-5 kDa for liposomes or nanoparticles 61

    . Although there are

    several studies evaluating the impact of the different lengths of PEG on liposomal

    formulations 62

    , PEG-2000 is the most commonly used 59,63

    .

    The amount of polymer has also been reported as another factor influencing certain

    characteristics of the liposomal formulations, because it can adopt two different spatial

    conformations. For PEG concentrations below 5-8%, the polymer has a mushroom-like

    conformation, whereas for higher concentrations, the most favorable conformation is

    brush-like, as is shown in Figure 4 6,64–66

    . The PEG in a mushroom structure exhibits a

    globular shape, overlapping with others, and hence, covering the nanoparticle surface 59

    . In

    contrast, the brush conformation, found for higher amounts, is provided by interactions

    between PEG chains, which show an elongated shape. Thus, in the case of percentages

    higher than 10%, PEG chains present lateral repulsion destabilizing the lipid bilayer 59

    .

    Moreover, it has been reported that the addition of higher PEG concentrations to the lipid

    mixture may promote a change in liposome structure. Thus, PEG concentrations around

    15% lead to a mixture of two different size populations 67

    , which correspond to liposomes

    together with micelles, also called “discoidal micelles” 68

    . Further, for PEG concentrations

    higher than 20%, liposomes disappear, significantly decreasing particle size and inducing

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    36

    the formation of spherical micelles 68

    . Thus, the standard condition used for liposomal

    formulations is 5% of PEG although it has been reported that 2% is sufficient to achieve a

    prolonged circulation. However, this factor should be optimized for each formulation,

    testing a range between 2-8% of polymer.

    The PEG structure can be also modified by the presence of the corona effect. This effect is

    due to the proteins that can attach to the PEG, extending or collapsing its structure and

    forming a shell as a corona, a process which is both very dynamic and complex 64–66

    . This

    phenomenon takes place very rapidly and can mediate the interactions between liposomes

    and their environment, strongly influencing the association with cell surface or even

    intracellular uptake 63,69

    . Therefore, a particular role is played by the PEG in

    immunoliposomes and targeted liposomes, in general, because their binding capability may

    be highly influenced by the type and amount of this polymer, as well as the corona effect 70

    and the nature of the ligand 50

    . Note that small ligand molecules can be embedded into the

    PEG layer 60,71

    or can display steric hindrances during the coupling step, shielding the

    recognition between the DSPE-PEG derivative ending and the ligand.

    However, despite its advantage for extending the circulation half-life and ligand coupling

    6,64,66, PEG may reduce cell interaction, endosomal escape, and drug release, leading to a

    decrease in the therapeutic drug index. This different behaviour at the pharmacokinetic and

    pharmacodynamic levels reflects the “PEG dilemma”, a contradictory effect associated

    with this polymer 72

    . To overcome this dilemma, the cleavable PEG molecules have been

    developed as a strategy able to release the polymer into the tumor tissue or even into the

    endosome. Most of these cleavable PEG derivatives are designed to respond to certain

    extracellular or intracellular microenvironmental conditions, such as acid pH, enzymatic

    activity, and others 72

    . Once the PEG is eliminated from the liposomes, they are able to

  • CHAPTER 1

    37

    interact with the tumor cell or the endosome. This strategy allows the enhancement of the

    antitumor activity of pegylated liposomes 72–74

    .

    Figure 4. Schematic representation of the PEG conformation. A) Brush conformation; B) Mushroom

    conformation.

    Nevertheless, clinical studies with pegylated nanoparticles have shown an unexpected

    immunogenic reaction, referred to as the Accelerated Blood Clearance (ABC)

    phenomenon, which is associated with repeated administrations 75

    . The mechanism is

    triggered by the first administration of pegylated liposomes. These accumulate in the

    spleen inducing the development of M immunoglobulins (IgM). With the second dose,

    these IgM may produce immune complexes, which are rapidly cleared, reducing the

    efficacy of the treatment 75,76

    . Currently, to reduce the impact of this ABC effect, several

    studies suggest that the dosing time for the second dose must be before the day 4 or later

    than the day 7 after the first dose 75

    . Interestingly, this effect does not occur for the third

    dose or for certain formulations encapsulating doxorubicin, where the ABC is decreased

    75.

    In sum, the development of immunoliposomes represents a complex process, where factors

    such as lipid composition, PEG amount, ligand coupling method, type of ligands and even

    encapsulated drug must be considered in order to obtain the most suitable formulation for

    attaining a specific desired effect 6,62,64

    .

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    38

    3. MECHANISM OF ACTION

    Target recognition in tumor cells by the ligands coupled to immunoliposomes results in the

    formation of a target-ligand complex. In general, this complex is internalized by

    endocytosis after multi-binding stimulation. This process is highly dependent on the nature

    of the ligand-receptor interaction 77,78

    , as well as the path followed by the formed

    endosome. This means that receptor-mediated internalization can either result in a clathrin-

    dependent endocytosis, as occurs for nanoparticles modified with Tf, leading to a lysosome

    formation, degrading, translocating or destabilizing the complex, that releases the drug into

    the cytosol and exerts both therapeutic and toxic effects 78

    , as is shown in Figure 5; or in

    caveolae-mediated endocytosis transport. This latter path allows immunoliposome

    internalization, bypassing lysosomes and increasing intracellular drug delivery.

    Thus, some liposomal formulations include endosomal escape lipids such as N-glutaryl-

    phosphatidylethanolamine (NGPE), DOPE/CHEMS or Listeriolysin O, that are able to

    disrupt the lysosome, releasing the cargo into the cytoplasm 79

    . Moreover, once the

    complex is destabilized, receptors can be recycled to the cell surface or degraded. Note that

    for certain growth factor receptors, their down-regulation involves in some cases an

    antitumor effect itself.

    For targeted liposomes, it is widely assumed that complex internalization must occur in

    order to achieve a higher anticancer effect than in the non-targeted approach. However,

    experimental evidence suggests that cellular internalization might not always be necessary,

    with cellular binding or interaction being sufficient to increase drug efficacy 55,56

    . Thus, in

    the case of liposomes targeted with non-internalizing ligands, after specific cell binding,

    their contents can be released in close proximity to other cells into the tumor

    microenvironment, with subsequent drug internalization, leading to cell death and localized

    damage 80

    .

  • CHAPTER 1

    39

    Figure 5. Schematic representation of the mechanism triggered by the complexes formed by the ligand-

    receptor interaction between targeted liposomes and the receptor on the tumor cell surface.

    This effect results in the increase of tumor antigens promoting an inflammatory process

    that might activate a favorable immunological response 81

    . Nevertheless, it has been

    demonstrated that, for immunoliposomes against internalizing epitopes, drug release is

    quicker than for non-internalizing ligands, leading to better and faster therapeutic efficacy

    24. To achieve a similar intracellular drug concentration, non-internalizing ligands require

    longer periods of time. Furthermore, on some occasions, those concentrations are not

    totally attained.

    Immunoliposomes are associated with favorable factors, such as the affinity or the strength

    of the interaction between the mAb and antigen, and the multivalency or ability of mAbs to

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    40

    attach to several antigen binding sites 82

    . This multivalency may be induced by the

    multimerization of antibodies or their fragments. This has important functional

    implications, because these ligands augment the valency and avidity of the complex,

    promoting a higher retention time on cell-surface receptors 83

    . Indeed, it has been reported

    that the use of multivalent antibodies to target liposomal formulations might also enhance

    the internalization of tumor antigens and other cellular signals, such as the induction of

    apoptosis and inhibition of cell growth 84,85

    .

    Another advantage for these immunoliposomes is that the internalization of encapsulated

    drugs contributes to overcome tumor therapy resistance bypassing efflux pumps like P-

    glycoproteins involved in the Multidrug Resistance effect (MDR) 7,86

    . One example is the

    transferrin targeted liposome encapsulating doxorubicin 55

    which was able to increase drug

    cytotoxicity 25 fold in resistant cells 87

    .

    4. PHARMACOKINETICS (PK) OF TARGETED LIPOSOMES

    Targeted liposomes improve the selectivity of the drug release into the tumor area,

    supporting an enhancement of drug efficacy 62

    . To achieve appropriate tumor drug

    concentrations, a prior evaluation of the pharmacokinetics (PK) of liposomal formulations

    is highly recommended 62

    .

    PK advantages have been reported for liposomal formulations in comparison with the free

    drug. Reduced body distribution, combined with a longer blood circulation time, in

    particular for pegylated liposomes, translates into a lower volume of distribution (Vd) and

    a higher Mean Residence Time (MRT) 6,56,88

    . PK information about drug exposure

    reflected in the half-life or area under the curve of drug plasma concentrations (AUC) is

    often reported during preclinical evaluation of different types of liposomal formulations in

    order to establish an increase in these parameters in relation to the free drug. However, it is

  • CHAPTER 1

    41

    difficult to perform an adequate comparison across formulations because PK properties are

    highly dependent on liposome characteristics and stability. In line with this, particle size

    and superficial charge play an important role. In the case of particle size, lower blood

    clearance is associated with < 30 nm. However, these nanoparticles show an inefficient

    tumor accumulation due to their higher renal excretion compared to bigger particle sizes 89

    .

    For liposomes with a particle size above 300 nm, these are mainly taken up by the liver and

    spleen 90

    . Therefore, the optimal range-size is between 80 and 150 nm 89,90

    . For particle

    charge, neutral liposomes present a lower RES effect compared to positively or negatively

    charged liposomes, triggering longer circulating times 91,92

    .

    Thus, body drug disposition is drastically modified after pegylated liposome

    administration, increasing the AUC, as occurs with Doxil. This presents 6 and 66 times

    higher AUC than non-PEG and the free drug, respectively 93

    .

    In general, this higher drug availability is assumed to correlate with an enhancement in

    tumor accumulation. There are many preclinical assays for studying these characteristics,

    although they involve the use of healthy animals, as can be observed in Table 4. Here, the

    differences between formulations are difficult to interpret due to the importance of the

    tumor during PK characterization 94,95

    .

    On the one hand, in animal tumor models, circulation half-life for targeted nanocarriers is,

    in general, shorter than for non-targeted carriers 96

    . This difference might be explained by

    the rapid receptor binding and complex internalization, which decrease the presence of the

    formulation in blood. An interesting example has been reported for EGFR targeted

    doxorubicin immunoliposomes administered to cancer patients. The half-life in blood was

    31 h, whereas for non-targeted it was 55-70 h 96

    . On the other hand, a linear PK has been

    reported for pegylated liposomes, but in some cases, this linear PK may be combined with

    a non-linear PK due to the specific binding to tumor cells. This is the case of liposomes of

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    42

    paclitaxel. The drug released from the formulation binds to plasma proteins in a linear and

    saturable manner 97

    .

    In other cases, stability is crucial because some liposomes present a biphasic plasma

    concentration-time profile, which is characterized by an initial rapid clearance due to the

    rapid drug release, followed by a slower elimination phase 93,98

    . To these aspects, it has to

    be added the lack of information regarding other important PK features, such as

    distinguishing between encapsulated and released drug, liposome tumor distribution, drug

    metabolites or immunogenic effects after repeated doses 93

    . In addition, particularly for

    immunoliposomes, ligand density is a factor directly related to their clearance rate as well

    as their ability to enhance tumor drug delivery. Thus, higher ligand density is correlated

    with higher elimination rate, as is observed in Table 5 99

    .

    Therefore, a balance between drug release and drug elimination would be required in order

    to obtain those concentrations associated with an adequate response. As a result, the

    pharmacokinetic and pharmacodynamic characterization of liposomal formulations should

    be applied to optimize the in vivo behaviour of the formulation and facilitate the translation

    of this information to patients 100

    . However, there are few studies in this regard.

  • Table 4. PK of different liposomal formulations IV administered to tumor-free animals.

    Type Formulation Encapsulated

    molecule

    EE

    (%)

    Particle

    size

    (nm)

    Animal

    model

    Dose

    (mg/kg)

    Cmax

    (mg/L)

    AUC

    last (h

    µg/mL)

    t1/2

    (h)

    CL

    (mL/h) Ref.

    LP Esphingomyelin: CH Vinblastine-N-

    oxide (CPD100)

    62.5 154

    Female SW

    30

    0.05 0.06 5.50 8.90 127

    LP

    Esphingomyelin: CH:

    DSPE-PEG 66.2 137 0.06

    0.17

    9.50 4.40

    LP DSPC: DSPEPEG2000:

    CH (TEA PN) Irinotecan

    100 110

    Female SD 10

    N.A.

    1407.80

    6.80 7.10

    128

    LP

    DSPC: DSPEPEG2000:

    CH (TEA SOS) 100 110

    2134.40

    10.70 4.69

    LP PC: CH Coumarin

    98 105

    Male SD 11.7

    4.89 2.32 0.41 1263.75 129

    T

    PC: CH: DSPEPEG2000:

    DSPEPEG2000-GAL 99 128 10.18 6.70 1.35 470.50

    LP EPC: DOPE: CH Gemcitabine

    15 177 Mice

    0.45

    N.A.

    9.58

    N.D. 54.30

    130

    T EPC: DOPE: CH 16 212 17.75 N.D. 26.70

    LP: non-targeted liposome; T: Peptide-targeted liposome; SD: Sprague-Dawley rats; SW: Swiss Webster mice; N.A.: non-available. The harmonization of PK

    parameters, some of them expressed by kg, has been done using the standard body weights, 25g for mice and 250g for rats.

  • Table 5. PK of different immunoliposomal formulations IV administered to tumor-free animals.

    Type Formulation Encapsulated

    molecule

    EE

    (%)

    Particle

    size (nm)

    Animal

    model

    Dose

    (mg/kg)

    Cmax

    (mg/L)

    AUC

    last (h

    µg/mL)

    t1/2

    (h)

    CL

    (mL/h) Ref.

    D Doxorubicin HCl

    Doxorubicin

    N.A.

    ---

    SD 5

    0.073 1.59 33.90 473.95

    131 LP Doxil® 109 44.81 569.30 14.70 1.85

    IL LP-Anti-CD147-DSPE-

    PEG-Mal 91 42.13 240.96 15.80 4.13

    D Daunorubicin

    Daunorubicin

    --- ---

    SD 4 N.A.

    7.40 1.72 107.75

    125

    LP S100PC: CH: mPEG2000-

    DSPE 93 105 269.0 11.19 3.75

    IL

    Anti-CD123-Mal-

    PEG2000-DSPE-

    S100PC: CH:

    mPEG2000-DSPE (1:400

    mAb/LP)

    91 113 114.3 7.61 8.00

    IL

    Anti-CD123-Mal-

    PEG2000-DSPE-

    S100PC: CH:

    mPEG2000-DSPE (1:800

    mAb/LP)

    90 109 160.3 9.84 6.00

    LP: non-targeted liposome; D: Active drug; IL: Immunoliposome; EE: Encapsulation efficacy; Cmax: Maximum serum concentration; AUC: Area under the curve; t1/2:

    Half life ; CL: Clearance; N.A.: non available.

  • CHAPTER 1

    45

    It is clear that differences across species may limit the translational application of these

    formulations. However, in an attempt to address this, more complex preclinical designs

    seem to be necessary to cover the relevant properties of these formulations. The application

    of physiological-based PK models which represent, in more mechanistic terms, liposomes

    and drug disposition in the different organs, in particular, the tumor, spleen, and liver, may

    help to yield preclinical and clinical findings and develop a better predictive model 97

    .

    5. CLINICAL TRIALS WITH TARGETED LIPOSOMES

    Despite the advantages reported for targeted liposomes, few formulations have reached

    clinical trials. In the present section, the status of the most promising targeted formulations,

    including immunoliposomes, will be discussed.

    5.1. Transferrin-targeted liposomes

    MBP-426,

    Transferrin (Tf) targeted liposomes encapsulating oxaliplatin, developed by Mebiopharm,

    have demonstrated a greater therapeutic effect than previous oxaliplatin formulations 16

    .

    This drug presents limited antitumor effects due to a PK limitation derived from its high

    levels of plasma proteins and erythrocyte binding that decrease free or therapeutic

    concentrations. To overcome this, several preclinical studies have reported a better PK and

    PD behavior following its encapsulation 48,106

    .

    Accordingly, the next step was liposome conjugation with Tf. This new formulation

    demonstrated higher tumor selectivity than non-targeted liposomes and, consequently,

    higher therapeutic activity. In a further optimization, lipid composition was modified by

    incorporating NGPE, which changes its conformation in an acid pH. This effect leads to a

    destabilization of both liposomes and lysosomes, promoting the intracellular release of

  • IMMUNOLIPOSOMES IN CLINICAL ONCOLOGY: STATE OF THE ART AND FUTURE

    PERSPECTIVES

    46

    oxaliplatin and