wp_clindev-immunotherapy_d03-final edits

16
White paper Seizing the future in oncology: Improving the clinical development of immunotherapies Brad Smith, Ph.D., Vice President of Translational Medicine, Quintiles Chris A. Learn, Ph.D., PMP, Senior Clinical Project Manager, Quintiles Eric Groves, MD, Ph.D., Vice President, Center for Integrated Drug Development, Quintiles Quintiles helped develop or commercialize 100% of the Top 50 best-selling Oncology products or compounds of 2013 Transform clinical development

Upload: chris-learn-phd-pmp

Post on 08-Apr-2017

234 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: WP_ClinDev-Immunotherapy_d03-final edits

White paper

Seizing the future in oncology:Improving the clinical development of immunotherapiesBrad Smith, Ph.D., Vice President of Translational Medicine, QuintilesChris A. Learn, Ph.D., PMP, Senior Clinical Project Manager, QuintilesEric Groves, MD, Ph.D., Vice President, Center for Integrated Drug Development, Quintiles

Quintiles helped develop or commercialize 100% of the

Top 50 best-selling Oncology products or compounds of 2013

Transform clinical development

Page 2: WP_ClinDev-Immunotherapy_d03-final edits

Introduction 3

Overview of the clinical immunotherapy landscape 4

Understanding who may benefit most from which approaches 6

Tumor-derived immune suppression reversal 6

Novel immunomodulators 7

Combinations 7

Adoptive T cell therapies 8

Cancer vaccines 8

Pharmacodynamic and predictive biomarkers in immunotherapies 8

Checkpoint inhibitor biomarker strategies 8

Biomarkers in immunotherapy development 10

Clinical development: goals and cautions 11

Goals for successful drug development 11

Cautions 11

Speculative issues 11

Evaluation of tumor shrinkage related endpoints 11

Management of autoimmune adverse events 11

Patient selection assays 12

Operational considerations before development 12

Key considerations about development 12

Current understanding 12

Conclusion 13

References 14

About the authors 15

Table of contents

Page 3: WP_ClinDev-Immunotherapy_d03-final edits

3 | www.quintiles.com

Introduction

Immunotherapy – using small molecules and biologics that provide therapeutic benefit by focusing the capabilities of the immune system on the tumor1 – promises to transform cancer care, having already shown striking patient responses. A recent Wall Street Journal article2 highlights “super-survivors” with exceptionally good responses to experimental immunotherapies. These individuals are described as transforming the world of oncology and charting new territory in both individual duration of survival and the potential for significant numbers of patients to benefit. As the article states, these results are reviving hopes that enlisting the power of the immune system may help to turn the tide against lethal and resistant forms of cancer.

Immunotherapy can be delivered in various therapeutic formats, with checkpoint inhibitors – which work by releasing the natural brakes on the immune system, enabling it to attack tumors – currently sparking the most interest. This represents an exciting and rapidly developing field, but understanding which patients will benefit most remains uncertain. It is not yet clear in all tumor types whether these drugs can be used to treat all patients or need to be targeted to selected populations, using precision medicine (with patient subgroups defined by molecular mechanisms), or personalized medicine (where an individual’s genetic profile is used to guide therapy). While these therapies do not produce universal or equal benefits, improvements in patient outcomes have been documented across many different cancer indications.

The human immune system is highly adaptive, and immunotherapies have the potential to greatly increase the patient’s leverage in fighting cancer. However, this potential requires the right setting and local environment cues. The immune system is an ideal anti-cancer agent, with large numbers of diverse types of immune cells (including 1018 T cells and 1022 antibodies); a high degree of specificity, with the ability to distinguish minute chemical alterations; and a long memory, with immunity lasting for up to several decades after effective antigen priming. This system has advantages for the patient and the prescribing physician in that the immune system attempts to target the vulnerabilities of the tumor in real time, rather than the prescriber having to use laboratory tests to hunt for vulnerabilities (which change over time) in the first place.

Among the immunomodulators, defined as medications used to help regulate or normalize the immune system, the immune checkpoint inhibitors are currently leading the way in clinical discovery and enthusiasm, given the exciting data yielded to date. Immune checkpoints refer to the inhibitory pathways that are hardwired into the immune system, and are involved in maintaining self-tolerance, as well as in modulating duration and amplitude of physiological immune responses in peripheral tissues to help minimize collateral tissue damage.4 Tumors take advantage of these systems or checkpoints to prevent the immune system from attacking and destroying the tumor.

Researchers are starting to elucidate the biology behind the anti-tumor immune response that is released by these checkpoint inhibitors, and have begun to clarify why certain patients and indications may be more amenable to this class of agents. Clearly, while the initial promise of immunotherapy rests largely in its aptitude for broad application in various patient populations, the algorithm for effective use in the oncology setting is excruciatingly nuanced, and reduces the reality of broader success at this time. The real hope now is to understand why immunotherapy can work, and when it cannot.

It should be noted that while checkpoint inhibitors dominate the current headlines in the clinical care communities, other promising approaches include novel molecular constructs such as chimeric antigen receptors (CARs), therapeutic combinations with old and new drugs, dosing regimen modifications, and vaccines. Older agents including IL-2 and the interferons remain in use, but may lose some visibility to these newer regimens. For researchers and oncologists alike, the most difficult algorithm to solve may not be how the cancer fights back, but rather how best to leverage the various components of the immune system armamentarium in this fight.

“Cancer immunotherapy refers to a number of approaches intended to activate the immune system to induce objective responses and disease stabilization.”– Drake, Lipson and Brahmer (2014)3

Page 4: WP_ClinDev-Immunotherapy_d03-final edits

4 | www.quintiles.com

Overview of the clinical immunotherapy landscape

While there is currently no conclusive evidence that immunotherapy will be a “magic bullet” as some may hope, there is reason for some confidence that this approach will be a dynamic and responsive treatment platform to fight cancer going forward. This therapeutic approach is undergoing a renaissance, since some of the more promising initial immunotherapy studies (for example, Rosenberg et al., 19945) involving IL-2 in the treatment of metastatic melanoma. The key to clinical efficacy will be to activate the immune system in a way that imparts high specificity to a target on the tumor or within its microenvironment (Figure 1).

Figure 1: Various types of cancer immunotherapy

Immunotherapy currently involves a wide range of cellular, humeral and other targets, as noted in Figure 1. As mentioned, the key question is how the various treatment options can be leveraged in the clinic to achieve real and durable benefits for patients. For example, basic research and clinical results suggest that certain combinations may induce new targets for the anti-tumor immune response. Options include potentially beneficial combinations of novel therapies and routine approaches such as radio- and chemotherapies. However, throughout the research and clinical care communities, as well as in biopharma companies, there are competing interests and viewpoints as to the best way to proceed.

Understandably, the most competitive arguments – and outcomes – will be those that are evidence-based and have the opportunity to be broadly applied. Some have argued that the notion of broad applicability is not viable, either in the context of precision or personalized medicine. Yet, if the requirement for specificity is inherent to the global response in the host (i.e., the immune system), then its applicability can be leveraged even when faced with overwhelming tumor diversity.

Products currently in development are listed in Figure 2 and include a large continuum of mechanisms and modalities. The immune system is built to recognize and distinguish self-antigens from non-self or foreign ones. As a result, the immune system is able to recognize self from altered-self, which is the case for cancer. While many cancers have circumvented endogenous anti-tumor immune responses, immunotherapy in its various forms has the potential to augment responses in order to mitigate tumor progression. Immunotherapy can operate along both lines of innate and adaptive responses.

Passive immunotherapy

Monoclonalanitbody

Conjugatedmonoclonal

antibody

Active immunotherapy

Oncolyticvirus

BiTE (Bi-specific

T cell Engager) mAb

Therapeuticvaccines

Cancer immunotherapy

Adoptive T celltechnology

Checkpointagents

Stimulatory(CD137, CD40, OX40)

Inhibitory(CTL40, PD1, LI, LAG-3)

TIL

CART

TCR

Source: Citi Research (Andrew S. Baum, Immunotherapy – The Beginning of the End for Cancer)

Immunotherapy is undergoing a renaissance, since some of the more promising initial studies involving IL-2 in the treatment of metastatic melanoma.

q800246
Highlight
delete "et al., 1994" so the parenthetical reads: (for example, Rosenberg 5) * with the 5 being superscript
Page 5: WP_ClinDev-Immunotherapy_d03-final edits

5 | www.quintiles.com

Notably, dendritic cells are antigen presenting cells that function at the intersection of innate and adaptive immunity. These are able to cross-present antigen to, and activate, T cells, making their development in the immunotherapy regimen especially important. As such, dendritic cells are a target of various immunotherapeutic approaches either through the use of adjuvant cytokines, which activate dendritic cells, or more directly through the use of dendritic cell vaccines. While the approaches currently in development described in Figure 2 are encouraging, a more complete understanding of the cellular and molecular components of the tumor-immune system interaction will be necessary for the development of rational and efficacious immunotherapies in the future.

Figure 2: Potential cancer immunotherapeutics under investigation (2015)

The current competitive landscape is highly complex, as shown in Figure 3, based on ongoing trials identified on clinicaltrials.gov. At present, competition is particularly strong in the areas of solid tumors, melanoma and non-small cell lung cancer. It remains to be seen whether classical tumor-type classifications will ultimately give way to molecular pathway-based classifications.

Figure 3: The competitive landscape

Checkpoint regulators Immunomodulators Combinatorial therapy Cancer vaccines

Negative effectorsPD-1PD-L1CTLA-4CD80KIRLAG-3TIM-3BTLAVISTA

Positive effectorsCD28OX40GITRCD27CD87HVEM

Negative effectorsTregsIL-4IL-10TGB-b

Positive effectorsIL-1IL-2IL-6IL-12GM-CSFIFN-gHLA-A*02

MPDL3280A +vemurafenib (Phase Ib)

Nivolumab + iliolumbar

Anti-LAG3 (BMS-986016) ± nivolumab

Nivolumab + ipilimumab

MEDI4736 + tremelimumab

MEDI4736 + dabrafenib + trametinib or trametinib alone

Pembrolizumab + pazopanib

Pidilizumab ± dendritic cell/RCC fusion cell vaccine

DC/APCsAGS-003DCVax®-LCvac

Peptides/protiensGV1001NeuVax™

StimuvaxRindopepimutPOP-103A

Virus vectorsPROSTVACCG0070INGN201TG4010

The immunotherapy landscape in 2015

Summary: Pipeline radar chart

Solid Tumors

Prostate Cancer

Pancreatic

Melanoma

NSCLC

RCC

Melanoma, NSCLC

Malignant Gliomas

Hodgkin Lymphoma, Myeloma, MDS, NHL

Hepatocellular

Glioblastoma

Gastric, SCLC, TNBC, HNC, Urothelial

Pembrolizumab (NSCLC)

BMS-936559

MED14736

Nivolumab + iliolumbar

MSB0010718CMED14736

MED14736 + tremelimumab

MPDL3280A + cobimetinib

MPDL3280A + bevacizumaband/or chemotherapy

Nivolumab

AMP-554

Nivolumab + interleukin-21

Anti-LAG3 (BMS-986016) ± nivolumab

Pidilizumab + sipuleucel-T +cyclophosphamide

Pidilizumab + gemcitabine

Nivolumab ± ipilimumab

Pembrolizumab

Pidilizumab

Pembrolizumab

Nivolumab

Nivolumab ± ipilimumab

Nivolumab ±ipilimumab

PembrolizumabPembrolizumab

MPDL3280A

Colon CancerSolid or Hematological Malignancies

PDL-192AMP-514 + MED14736

MPDL3280A + vemurafenib (Phase Ib)

Nivolumab + ipilimumab

Combination

Mono therapy

Nivolumab + multiple class 1 peptides & montanide ISA 51 VG

MED14736 + dabrafenib + trametinib or trametinib alone

Pembrolizumab

Pembrolizumab

MPDL3280A ± bevacizumab vs sunitinib

Nivolumab + ipilimumab

Nivolumab ± ipilimumab Nivolumab

Nivolumab sequentially with ipilimumab

MPDL3280A + erlotinib (Phase lb)

MPDI4736 + tremelimumab

Nivolumab ± gemcitabine/cisplatin,pemetrexed/cisplatin, carboplatin/paclitaxel,bevacizumab, erlotinib, ipilimumab

Nivolumab

MPDL3280A

MPDL3280A

Pembrolizumab

Advanced Solid Tumors

Nivolumab

MPDL3280A

Nivolumab

Pembrolizumab(Phase II/III)

Pidilizumab ± dendritic cell/RCC fusion cell vaccine

Nivolumab

Pembrolizumab + pazopanib

Nivolumab + sunitinib, pazopanib, or ipilimumab

Phase I

Phase II

Phase III

Prepared by ESD Competitive Intelligence & Response. Only ongoing trials have been considered for analysis.

q800246
Highlight
delete
q800246
Highlight
add date to read: Figure 3: The competitive landscape, 2014
Page 6: WP_ClinDev-Immunotherapy_d03-final edits

6 | www.quintiles.com

While the competitive landscape is crowded and promising, the most important aspect to consider is which of these agents in development will help to address the most fundamental limitations in oncology clinical care. Among the notable current challenges in the oncology clinic, the following considerations will largely determine the future success of these competitors:

• Understanding complex immune mechanisms of action and correctly applying therapy

• Determining and utilizing clinically relevant biomarkers to characterize immune response and identify the baseline patient anti-tumor status

• Defining optimal dose and schedule/sequence especially in combination settings

• Patient selection in the absence of clear selection criteria or targeted oncogenic drivers

• Trial design in the context of optimal dose, schedule/sequence and delayed response

• Leveraging increased efficacy versus increased toxicity

• Defining meaningful and standardized clinical endpoints for these classes of agents

Of greatest importance is the fact that the immune system does not operate separately from other biological systems in the patient, including the inflammatory system. Thus, rationally selected targets and targeted patient groups must be defined upfront. To avoid wasted time and money, an informed approach is needed for the immunotherapy platform from the outset of development. The rapidly emerging research results describing immunotherapy targets, drug combinations and responsive patient characteristics should lead to better informed studies and more successful trial outcomes.

Understanding who may benefit most from which approachesTargeting the right therapy to the right patient is likely to become a central goal of immunotherapy. While classic cytotoxic chemotherapy does have tumor type specificity, refinements of this targeting have been elusive. More refined targeting has come from identification of oncogenic drivers that can be targeted by small molecules or antibodies, with examples including Herceptin,® Rituxan,® and BRAF and ALK inhibitors. Here, the target is identified by the researcher, but, as noted earlier, there are limitations in knowledge of tumor biology and biomarker technology that limit efficacious application for many potential targeted therapies. However, the recent success of some small molecules targeting oncogenic drivers has been dramatic.

It should be noted that immunotherapy follows a new paradigm in tumor targeting. Instead of specifically inhibiting the impact of an oncogenic driver, the immune response mounts a powerful, multi-facetted anti-tumor attack, which is characterized by specificity. This is clearly the case for immunomodulatory approaches and is also true for tumor vaccines. The immune system is believed to have potential to be more successful in identifying tumor vulnerabilities than prior approaches and to have the potential for wide application. However, this new paradigm presents new complexity and uncertainty in identifying and utilizing a precision medicine approach.

Tumor-derived immune suppression reversalA single tumor can employ a cadre of mechanisms to overcome immune responses and orchestrate systemic disease, despite the expanse and specificity brought forth by both innate and adaptive immune mechanisms. Tumor-derived immune suppression involves various pathways, such as:

• Immune evasion mechanisms that may involve downregulatory cytokines (TGF-β, IL-4, IL-6, IL-10), immunosuppressive cells (regulatory T cells [T-regs], macrophages), or altered immune activation (loss of major histocompatibility complex [MHC] receptors, indoleamine 2,3- dioxygenase [IDO] production)

• Immune dysregulation mechanisms that include: inactivation of or failure to activate tumor-specific T cells; insufficient antigen processing and/or presentation; ineffective clonal priming or homing; disruption of effector cell maturation or expansion; or T-cell anergy, a proposed mechanism of immunologic self-tolerance in which T cells become functionally inactivated after previous stimulation6

Since the immune system does not operate separately from other biological systems in the patient, including the inflammatory system, rationally selected targets and targeted patient groups must be defined upfront.

Page 7: WP_ClinDev-Immunotherapy_d03-final edits

7 | www.quintiles.com

• Immune editing by developing tumors to shape, altering their appearance to the immune system

• “Non-inflamed” tumors, which are defined as cancer without immune infiltrates or other characteristics of an innate anti-tumor response.

Novel immunomodulatorsImmunotherapeutic approaches to cancer involve extensive and sophisticated ligand-receptor interactions at the cellular level, a veritable ”checkpoint chess match,” as illustrated in Figure 4. This figure shows co-stimulatory and co-inhibitory interactions between a T cell and a dendritic cell, a tumor cell, and a macrophage, respectively, in the tumor microenvironment.7 A reasonable question for the researcher to determine may be which of these interactions is most crucial to either host or tumor advantage. A seemingly less reasonable answer may be that all of these interactions are important, and furthermore, there is a dynamic gradient across which this importance fluctuates.

So, while the concept of checkpoint inhibition is encouraging and provocative in terms of therapeutic promise, the understanding of this process is far from complete. It is likely that an integrated and complex set of interactions are involved (Figure 5).3 However, this complexity also leads to many new drug targets and combinations. The early results from the various checkpoint inhibitors currently in trials and the few combinations of checkpoint inhibitors suggest very promising synergies for future therapeutic strategies.

CombinationsElaboration of basic innate and adaptive tumor immunity research has improved the potential for developing efficacious treatments. Clinical evaluation of multiple regimens used in concert has gained increased acceptance and traction over the past decade. This has included the combinatorial use of cancer vaccines, monoclonal antibodies, recombinant cytokines, and adoptive cellular infusions, all with or without some level of conventional chemotherapy and/or radiotherapy in an adjuvant setting. Yet, while these combinations increase tumor immunity in many patients, the majority still succumb to progressive disease. As such, some investigations have highlighted efficient dendritic cell activation and inhibition of negative immune regulation as central pathways for intervention. Early-stage clinical testing raises the possibility that combinatorial approaches that augment tumor antigen presentation and antagonize negative immune regulation may accomplish significant tumor destruction without the induction of serious autoimmune disease.

Dendriticcell

Tumor

T cell

TCR

MHCPD-L1

PD-1

PD-1

PD-L2MHC-II

LAG-3

Tissuemacrophage

TIM-3

PD-1

TCR

MHC

PD-L1

Galectin 9

OX404.1BB

CTLA-4

CD28

TCRCD40L

PD-1PD-1

ICOS

GITR

4.1BBL

PD-L1PD-L2

CD40

MHC

CD80

CD86

ICOSL

OX40LGITRL

Tumor cell or antigen-presenting cell

Tumor-specificT-cell

PD-1

CTLA-4

Anti-CTLA-4CD28T-cellreceptor

Antigen

MHC

B7PD-L1

Anti-PD-1

Figure 4: Ligand-receptor interactions between cells in the tumor microenvironment

Figure 5: Immune checkpoint blockade

Page 8: WP_ClinDev-Immunotherapy_d03-final edits

8 | www.quintiles.com

Adoptive T cell therapies Adoptive T cell therapy begins with ex vivo stimulation of lymphocytes in a non-tolerizing environment, which is then followed by re-infusion of activated T cells into patients for the purposes of measurable, clinically relevant anti-tumor outcomes. There are varying sources and types of T cells used for adoptive therapy, which include tumor infiltrating lymphocytes (TILs), T cells engineered to express a cancer-specific TCR, and most notably T cells engineered to express a chimeric antigen receptor (CAR), which combines the extracellular portion of an antibody with the T cell receptor signaling machinery. Of these approaches, CAR T cells, with uniform specificity, appear to be especially promising, demonstrating at times considerable clinical efficacy and durable responses, but are technically limited to some degree by transduction efficiency and potential toxicity. At this time, further research and successful applications are needed to warrant long-term continued development.

Cancer vaccinesCancer vaccines have also shown promise based on advances in adjuvants, combinations, biomarkers and new targets. These vaccines, which contain tumor cells or antigens, stimulate the patient’s immune system to produce orchestrated cellular and/or humeral responses that destroy tumors and, in theory, prevent relapses of the cancer. However, unlike vaccines for other disease that prevent the occurrence of the disease in the first place, there is not a vaccine in development that can prevent the onset of cancer. Cancer vaccines are used primarily as a treatment after diagnosis, and this may account for their limited clinical utility to date. Nevertheless, a variety of vaccination platforms have been put into clinical development, including antigen vaccines, tumor cell vaccines, anti-idiotype antibody-based vaccines, dendritic cell vaccines, DNA vaccines and viral-vector based vaccines.

Pharmacodynamic and predictive biomarkers in immunotherapiesThe immune system and tumor biology present many biomarker options. For the immune system, immune and inflammatory status and response to therapy can be measured. For the tumor, metrics include the tumor profile and response to therapy, including resistance mechanisms. Other metrics include: lymphocyte profiling (using flow cytometry), serum factors (using immunoassays), infiltrating cells (immunohistochemistry), gene expression (array-based GEP or RNA-sequencing), human leukocyte antigen (HLA) classification (multiplex polymerase chain reaction [PCR]), and T cell repertoire (NGS).

Checkpoint inhibitor biomarker strategiesThese typically focus on target expression such as PD-L1 immunohistochemistry or biomarkers reflecting anti-tumor immune response such as tumor infiltrating lymphocytes, serum factors or immune cell profiles and spectrum of genetic abnormalities in the target tumor.

Recent literature suggests that the number of somatic mutations or mutation load may also predict response. This idea emerged when a correlation between mutation load and patient response was first noted.8 Reports in the New England Journal of Medicine and Nature 9,10,11 have described how patients with many tumor mutations provide neo-antigens to the immune system that can be processed into antigen peptides, resulting in a T cell response. These responses may be followed by various NGS or mass spectrometry-based methods to characterize specific immunogenic peptides, T cell repertoires and transcriptome response signatures. Not only can an immune response to the neo-antigens be identified, but common antigenic peptides have been identified that are shared with antigens found in infectious disease. The size of the T cell repertoire or the clonal diversity also reflects the immune response and correlates with patient response to the therapy. Response signatures using the transcriptome or gene expression profile have also been identified, promising to improve understanding of responders and non-responders, and to predict response.

Cancer vaccines stimulate the patient’s immune system to produce orchestrated cellular and/or humeral responses that destroy tumors and, in theory, prevent relapses of the cancer.

Page 9: WP_ClinDev-Immunotherapy_d03-final edits

9 | www.quintiles.com

Examples of pharmacodynamic and predictive biomarkers for proof of concept and approval are:

• Pharmacodynamic biomarker endpoints for demonstration of target engagement and biological activity, including: target engagement and dose, receptor occupancy, biological activity, lymphocyte profile and serum factor panel, TILs, standardization and reproducibility; feasibility of pre- and post- sample collection; quantification of TILs; immunohistochemistry or multiplex immunofluorescence (IF); and digital pathology scoring.

• Predictive biomarker endpoints include PD-L1 expression, which may be of limited value, especially in combination studies; TILs, lymphocyte profile, gene expression, and potentially, inflammatory signature; T cell repertoire NGS profiling, which may have potential for implementation in global studies; somatic mutation profiling and combination studies; and translation of early correlations to robust CTAs and future companion diagnostic tests.

Checkpoint negativity can be an unreliable biomarker, placing into question how useful it will be going forward. There are several reasons for this. First, assays are technically imperfect, and results may differ depending on the antibody/assay (tumor vs. immune cells). Second, there may be variable expression, tumor heterogeneity, and inducible gene sampling error (false negative). Third, archived tissue may provide a less predictive test result than recently biopsied tissue. As a result, checkpoint negativity may be more useful in determining which tumors (rather than which patients) to treat. Expression may be less relevant for combination therapies as the expression of PD-L1 and possibly other immunotherapy targets are often impacted by targeted agents as well as radiation or chemotherapy.

In biomarker strategies for cancer vaccines, critical issues to be addressed include the patient’s immune status and ability to mount an immune response, and whether the immune response predicts patient outcomes. Two major goals in a cancer vaccine strategy are the ability to determine immune response, and to identify biomarkers that are predictive of immune response and patient outcome. Candidate biomarkers include target antigen expression, tumor burden, lymphocyte or serum factor inflammatory profile, TILs, antigen-specific immune response, predictive gene expression signatures and mutation load. However, to date, no successful single biomarker or approach has been confirmed.

The question remains as to whether there will be a need for predictive biomarkers for immunotherapies. Examples of complexities in this area are broad responses in many tumor types, the fact that combination therapies induce targets, responses are observed in biomarker negative patients, and complete responses are observed with checkpoint inhibitors while cancer vaccines lack candidate predictive markers. With pharmacodynamic analysis, biological activity is easily demonstrated, yet complexity is not captured. Safety biomarkers reflecting autoinflammatory response may also be important, and biomarkers may be useful in evaluating resistance where mechanisms are unknown. Early results examining resistance mechanisms to CTLA4 treatment in melanoma indicate that expression of parallel checkpoint proteins may be one mechanism that could be determined in a biomarker testing strategy.

Checkpoint negativity can be an unreliable biomarker, placing into question how useful it will be going forward.

Page 10: WP_ClinDev-Immunotherapy_d03-final edits

10 | www.quintiles.com

Biomarkers in immunotherapy development

Biomarkers and related technologies offer potential strategies to support drug development and patient care including approaches that include immunotherapies.

A biomarker is a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or biological responses to a therapeutic intervention

– National Institutes of Health (NIH)12

Biomarker classification and utility: supporting clinical study objectives and endpoints

Biomarkers offer two types of information that can support clinical study objectives and endpoints: pharmacodynamic, revealing target engagement and the magnitude of biological response post therapeutic intervention; and predictive, identifying patients who are susceptible to a particular drug effect, which may involve benefit or harm.

Biomarkers can also provide markers of drug efficacy, toxicity or resistance. In addition, they may be useful in pharmacodynamics-based therapeutic drug monitoring, as surrogate endpoints (validated indicators of future clinical outcome), or as non-surrogate endpoints (unvalidated indicators of future clinical outcomes).

In patient care, biomarkers may be diagnostic, indicating the presence or absence of pathogenic processes, or prognostic, providing information on the likely future clinical course in the absence of a therapeutic intervention. Used in companion diagnostics, biomarkers help predict responses to therapy. They may also be used in multiplex or multi-analyte diagnostic panels, in next-generation sequencing (NGS) genomic panels, or in whole exon sequencing (WES) or whole genome sequencing (WGS).

The complexity of the immune system and tumor biology also results in a wealth of biomarkers and potential targeted indications and patient populations. New diagnostic tools, such as more powerful and better standardized flow cytometry and next-generation sequencing, can better profile immune response and utilize the immune repertoire for patient selection. Application of these tools in clinical development and patient care has yet to be fully exploited. Optimal clinical strategies, including drug dose and schedule to minimize risk of toxicity and the most appropriate clinical endpoints, must address the unique mechanisms of action of these drugs.

As the field moves beyond initial successes, it will be important to explore these biomarker opportunities as well as address the critical questions in clinical trial design to best advance immuno-oncology clinical research.

Page 11: WP_ClinDev-Immunotherapy_d03-final edits

11 | www.quintiles.com

Clinical development: goals and cautions

Goals for successful drug developmentThe process for developing immunotherapy drugs follows the same steps as other biopharma products, including establishing safety, efficacy, dose and schedule, target indication and patient population, and providing support for clinical and reimbursement aspects of the target product profile. Progress to date in each of these areas for tumor vaccines, immunomodulators, cell-based therapies and combinations is illustrated in Figure 6.

Figure 6: Goals for successful drug development: Progress to date

Goal Tumor vaccine Immuno-modulator Cell based Combinations

Dose and schedule (fed/fasting)

Relationship to efficacy difficult

MTD and PK/blood concentrations

Empiric Ill defined

Target indication/patient selection

Defined by vaccine antigens

Appears broad but empiric

Defined by MOA Empiric

Registration efficacy data

Has been problematic Exciting Unknown Exciting

Adequate toxicity profile Usually benignRanges from tolerable to challenging

Ranges from tolerable to challenging

Likely challenging

Support for the clinical and reimbursement aspects of the TPP

Requires attention Requires attention Requires attention Requires attention

Supporting: Pop-PK, Special populations, Companion dianostic support

Requires attention Requires attention Requires attention Requires attention

CautionsPotential pitfalls in the development of new products in this area include failure to anticipate unique properties of the response to the immunomodulator. For example, the mechanism-of-action (MOA) model may be unrepresentative, leading to targeting of unresponsive subpopulations; patient selection assays may be weak; classic endpoints can be problematic; combinations of immunomodulators may lead to unexpected autoimmune syndromes, and may interfere with desired immunomodulatory effects in ways not seen in animal models. There may also be issues relating to accrual of patients in the target subpopulation (due to requirements for a novel screening process, or to the small size of the subpopulation). Monitoring of pharmacodynamic (PD) responses can also pose challenges related to sample handling, lack of availability of clear surrogates of desired bioactivity, or the fact that relevant samples may only be obtainable from repeated tumor biopsies.

Speculative issuesOther issues that may pose challenges are the potential for HLA-related ineffective immunodominance or novel adverse events (AEs), and the fact that repeated use of cytotoxic therapies may eliminate the relevant immunologic response. Unanticipated immune escape mechanisms may also occur, and promising immunomodulators may fail because of the need for multivariate patient selection assay systems.

Evaluation of tumor shrinkage related endpointsEndpoints related to tumor size can be complicated by the potential for delayed tumor shrinkage following therapy, or for flare before shrinkage. Here, training of investigators and response evaluators is essential.

Management of autoimmune adverse eventsTo date, vaccines have not been limited by AEs, but cellular therapy side effects have not been well characterized. For immunomodulators, there may be serious AEs. For example, high dose IL-2 may be associated with significant capillary leak with potential intensive care requirement; CTLA-4 inhibitors have potential for multiple autoimmune AEs; PD-1 inhibitors are better tolerated but may be linked to pneumonitis,

Page 12: WP_ClinDev-Immunotherapy_d03-final edits

12 | www.quintiles.com

immune mediated colitis, hepatitis, hypophysitis, nephritis (and renal failure), hypo/hyperthyroidism and other adverse reactions. Combinations of CTLA-4 and PD-1 look clinically promising but toxicity management is a challenge. There is a need to anticipate the need to develop official guidance for patient management in these areas.

Patient selection assaysCurrent assays for patient selection may not always have adequate specificity and sensitivity. Researchers face the question of whether to raise the threshold to assure more specificity, a move that might exclude some potential responders. Another issue is setting a price point for a new product in an environment where the target population may shrink due changes in the approach to patient selection. Current assays may require patient tumor biopsies, driving a need to focus on patient selection assays using blood or urine.

Operational considerations before developmentKey operational considerations before starting development of a new immunotherapeutic include the need to:

• Fully understand the clinical question to be answered• Appreciate the value proposition of the drug that is being developed• Employ dedicated teams with operational expertise and vision• Focus on achieving simplified interpretations in complex systems• Apply rigor in graduating the drug to the next phase of development• Ask the tough questions to address qualifications that must be met.

Key considerations about developmentRegulatory agencies, including the U.S. Food and Drug Administration and European Medicines Agency, are showing significant interest in the potential of immunotherapy. When antitumor activity is high, regulators typically value objective response rate (ORR) over progression-free survival (PFS) in single-arm trials. Controlled Phase 3 studies will remain extremely important here, because they take into account prognostic factors. Post-marketing safety monitoring is particularly critical, due to the low patient numbers treated in an accelerated drug development model.

Current understandingCurrently available information indicates that targeted small molecule therapy can yield high response rates, but is often prone to resistance, while immunotherapy can produce durable antitumor responses in some patients with cancer. Treatment of patients with immune checkpoint inhibitors can differ from use of conventional therapies in the need to identify unconventional responses, and to understand and manage immune-related adverse events. Accumulating evidence indicates that checkpoint inhibitors are active in multiple tumor types, yet clinical management and safety monitoring remain key for therapeutic success. Drug sequencing may be critical to the design and implementation of complex drug combinations, and ongoing studies will help define the optimal use of checkpoint inhibitors in different tumor types as single agents or as part of combination therapy.

Regulatory agencies, including the U.S. Food and Drug Administration and European Medicines Agency, are showing significant interest in the potential of immunotherapy.

Page 13: WP_ClinDev-Immunotherapy_d03-final edits

13 | www.quintiles.com

Conclusion

The landscape for immunotherapies in oncology is expected to expand greatly in the near future as the initial checkpoint inhibitors come to market, while development continues on a broad set of immunomodulators and cancer vaccines. Follow-up checkpoint inhibitors targeting LAG3, OX40, IDO1 and other novel proteins and novel combinations may provide new options for cancer patients.

This is an area with many potentially competing products, and a need for these to be differentiated from one another. Results to date provide a new understanding of the mechanisms of action of immunotherapies and provide predictive approaches for selecting indications and patients. It remains to be seen how this information may be used to differentiate immunotherapy drugs and aid clinicians in their decision-making.

For clinical trials, the overarching goal remains to achieve quick and efficient immunotherapy studies that yield high-quality data, and accelerate delivery of much-needed, new cancer therapies to patients. The trials must also provide a greater understanding of which patients respond and why. These approaches provide more evidence that cancer is a genetic disease that is translated through proteins, peptides and cellular mechanisms into sickness or cures. Immunotherapies are proving to be a very powerful approach to attack these disease-related alterations, leading to significant patient benefit.

Page 14: WP_ClinDev-Immunotherapy_d03-final edits

14 | www.quintiles.com

References

1. Vatsan RS, Bross PF, Liu K, Theoret M, De Claro AR, Lu J, et al. Regulation of immunotherapeutic products for cancer and FDA’s role in product development and clinical evaluation. Journal for ImmunoTherapy of Cancer 2013, 1:5 doi:10.1186/2051-1426-1-5; http://www.immunotherapyofcancer.org/content/1/1/5

2. Winslow R. Cancer’s Super-Survivors: How the Promise of Immunotherapy Is Transforming Oncology. Wall Street Journal, December 14, 2014.

3. Drake CG, Lipson EJ, Brahmer JR. Breathing new life into immunotherapy: review of melanoma, lung and kidney cancer. Nat Rev Clin Oncol. Jan 2014; 11(1): 24–37. Published online Nov 19, 2013. doi: 10.1038/nrclinonc.2013.208 http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4086654/

4. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews Cancer 12, 252-264 (April 2012) | doi:10.1038/nrc3239.

5. Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA. 1994 Mar 23-30;271(12):907-13.

6. LaSalle JM, Hafler DA. T cell anergy. FASEB J. 1994 Jun; 8(9): 601-8.

7. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. 2013 Oct 1;19(19):5300-9. doi: 10.1158/1078-0432.CCR-13-0143

8. Champiat S, Ferté C, Lebel-Binay S, Eggermont A, Soria JC. Exomics and immunogenics: Bridging mutational load and immune checkpoints efficacy. Oncoimmunology. 2014 Jan 1;3(1):e27817. Epub 2014 Jan 16. PubMed PMID: 24605269; PubMed Central PMCID: PMC3937193.

9. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014 Dec 4;371(23):2189-99. doi: 10.1056/NEJMoa1406498. Epub 2014 Nov 19. PubMed PMID:25409260.

10. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014 Nov 27;515(7528):568-71. doi: 10.1038/nature13954. PubMed PMID: 25428505; PubMed Central PMCID: PMC4246418.

11. Yadav M, Jhunjhunwala S, Phung QT, Lupardus P, Tanguay J, Bumbaca S, et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature. 2014 Nov 27;515(7528):572-6. doi: 10.1038/nature14001. PubMed PMID: 25428506.

12. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clinical Pharmacology and Therapeutics. 2001;69(3):89–95.

Page 15: WP_ClinDev-Immunotherapy_d03-final edits

15 | www.quintiles.com

About the authors

Brad Smith, Ph.D.Vice President of Translational Medicine, QuintilesBrad Smith currently is a Vice President of Translational Medicine within the Integrated Clinical Services Group at Quintiles. In this position, Brad supports laboratory, clinical and diagnostic strategies for drug development as well as the development of innovative tools for targeted drugs and companion diagnostics. Previously, Brad led Corporate Development at Cell Signaling Technology, an innovative biotechnology company in the life sciences field. In this position, he focused on new diagnostic and clinical partnerships and markets. His previous positions at Cell Signaling Technology include management of research and clinical technology development departments and laboratories. Previous to Cell Signaling Technology, Brad directed product development and production at Santa Cruz Biotechnology, helping to build that company into one of the largest supplier of research tools for basic research. Brad’s scientific background includes research positions at Stanford University and University of California, San Francisco focused on cellular signaling mechanisms of disease. He holds a Doctoral degree from Stanford University and Master’s and Bachelor’s degrees from University of California, Santa Cruz.

Chris Learn, Ph.D., PMPSenior Clinical Program Manager, Oncology, QuintilesChris Learn, Ph.D, PMP, is Senior Clinical Program Manager, Oncology, Quintiles. He has over 10 years of experience leading investigator led oncology trials in academic settings and in industry. His expertise includes the development of molecular immunotherapies for malignant glioma. Prior to joining Quintiles, he held senior positions in clinical research at Surgical Review Corporation, The Hamner Institutes for Health Sciences and Duke University Medical Center.

Eric Groves, M.D., Ph.D.Vice President, Center for Integrated Drug Development, QuintilesBoard certified in oncology and internal medicine, Dr. Groves has more than 20 years’ experience in drug development as corporate officer/senior manager, clinician and researcher. Prior to joining Quintiles in August of 2007, Dr. Groves was at Ligand Pharmaceuticals Inc., starting in August 1999 as Vice President, Project Management and corporate officer. From 1994 until joining Ligand, Dr. Groves held a number of positions at Sanofi Pharmaceuticals, most recently as Vice President, Project Direction, where he was responsible for the worldwide strategy of and project direction for late-stage Sanofi oncology projects. From May 1991 through October 1994, Dr. Groves served as Senior Project Director for the research division of Sterling Winthrop Corporation, and served as acting Vice President, Discovery and Clinical Research, Immunoconjugate Division. He was Director of Clinical Research and Development at CETUS Corporation from 1989 through 1991.

Page 16: WP_ClinDev-Immunotherapy_d03-final edits

Cop

yrig

ht ©

201

5 Q

uint

iles.

All

right

s re

serv

ed.

15.0

035-

1-05

.15

Contact usToll free: 1 866 267 4479 Direct: +1 973 850 7571Website: www.quintiles.com Email: [email protected]