Review
Clinical deployment of antibodies for treatment of melanoma

https://doi.org/10.1016/j.molimm.2015.01.025Get rights and content

Highlights

  • Melanoma treatment has been transformed using T-cell checkpoint antibodies.

  • Antibodies to CTLA-4 and PD-1 have had the largest impact on melanoma management.

  • Combination T-cell checkpoint therapy holds great promise for clinical development.

  • OX40 and 4-1BB are T cell costimulators with clinical potential in melanoma.

Abstract

The concept of using immunotherapy to treat melanoma has existed for decades. The rationale comes from the knowledge that many patients with melanoma have endogenous immune responses against their tumor cells and clinically meaningful tumor regression can be achieved in a minority of patients using cytokines such as interleukin-2 and adoptive cellular therapy. In the last 5 years there has been a revolution in the clinical management of melanoma in large measure based on the development of antibodies that influence T cell regulatory pathways by overcoming checkpoint inhibition and providing co-stimulation, either of which results in significantly more effective immune-mediated tumor destruction. This review will describe the pre-clinical and clinical application of antagonistic antibodies targeting the T-cell checkpoints cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1), and agonistic antibodies targeting the costimulatory pathways OX40 and 4-1BB. Recent progress and opportunities for future investigation of combination antibody therapy will be described.

Introduction

The humoral immune system is capable of making antibodies diverse enough to recognize over 10 billion foreign antigens with targets as diverse as microbial pathogens and tumor cells. After binding to antigen, antibody effector function is mediated by the following: complement fixation, Fc receptor binding leading to degranulation of neutrophils, engagement of other immune cells with cytotoxic function, antibody-dependent cellular cytotoxicity (ADCC) or prevention of binding of the antigen to adhesion or signaling molecules. These events in turn can promote a variety of regulatory functions that modulate the immune response including immunoglobulin class switching, cytokine release, B-cell memory and feedback regulation that influences immune enhancement or suppression. The adaptability and diversity of this system is carefully regulated, and B cells that produce antibodies that bind to self-antigen are eliminated. Autoimmune disease with significant clinical consequences can occur in patients where there is failure to eliminate or regulate self-reactive B cells. One could conjecture that allo-antibodies with the ability to promote immunologic memory or cytotoxic function in the setting of chronic infection or malignancy by engaging stimulatory T-cell receptors or inhibiting T-cell check-point proteins might confer some evolutionary benefit, but no naturally occurring antibody has been described with these properties.

Therapeutic antibodies have been used in medical care and research for decades, but in the last 15 years they have become commonplace in oncologic management. The majority of these monoclonal antibodies are antagonistic, and were engineered to block a protein antigen of interest or to induce ADCC. The value of this approach has been translated numerous times in clinical medicine. Some examples from the last 15 years in medical oncology include rituximab (used in CD20+ B-cell malignancies) (Davis et al., 2000), trastuzumab (erb-b2-overexpressing breast cancer) (Slamon et al., 2001) and cetuximab (tumors that have mutated EGFR, such as colon cancer and head and neck cancer) (Baselga et al., 2005, Cunningham et al., 2004). The mechanism of tumor elimination with these therapeutic agents is complex and likely includes antibody-dependent cellular cytotoxicity (ADCC) as well as inhibition of growth pathways salient to tumor growth (Arnould et al., 2006, Ferris et al., 2010).

Another area of therapeutic investigation has been the use of antibodies as immunomodulators. Anti-CD3, an antibody that binds the CD3 component of the T-cell receptor (TCR), administered at high doses causes immunosuppression and can be used to treat allograft rejection in patients who have received solid organ or bone marrow transplants (Bluestone et al., 1993). Anti-CD3 at low doses has immune stimulating effects and can trigger T-cell activation through the zeta chain of the TCR (Urba et al., 1992). Although anti-CD3 is not currently used as an anti-cancer agent, the observation that a T-cell-directed antibody could trigger T-cell cytotoxicity was an important conceptual insight that has lead to a proliferation of research on antibodies that influence the activation state and behavior of T cells resulting in enhanced anti-tumor immune responses. The CD3-zeta chain component has been used as a component of chimeric antigen receptors (CARs) to target tumor antigens with remarkable clinical activity in acute myelogenous leukemia and may have application in a wide variety of malignancies (Mardiros et al., 2013). Bispecific antibodies that link VH and VL for CD3 binding with the VH and VL that binds a tumor-associated antigen (e.g. CD19) have been used to redirect T cells to kill tumor cells. This approach has been studied in hematological malignancy and has recently garnered FDA approval, (Topp et al., 2014) but could also be applied to other solid tumors including melanoma.

A rapidly expanding knowledge of the receptors and pathways that regulate T cells, natural killer (NK) cells and antigen-presenting cells (APC) has identified the targets to which the current generation of melanoma therapeutic antibodies has been engineered. The T-cell pathways that have been most extensively studied to develop therapeutic antibodies in cancer are the T-cell checkpoints known as cytotoxic T lymphocyte antigen-4 (CTLA-4 designated CD152) and programmed death-1 (PD1 designated CD279). In addition, the study of the costimulatory pathways OX40 (CD134) and 4-1BB (CD137) may also yield therapeutic advances (Fig. 1). This review will describe the T-cell pathways and therapeutic antibodies that are transforming the care of patients with melanoma and other cancers, the rationale for combination antibody therapies currently undergoing clinical investigation and future questions that need to be answered to optimize antibody-based cancer therapy. Although the pathways will be presented individually, it is important to recognize that they function concurrently and that the final effect on T-cell activation is the result of the integration of the multiple coinhibitory and costimulatory signals.

Section snippets

CTLA-4: Pre-clinical observations

The steps that lead to T-cell activation include peptide antigen presentation by an antigen presenting cell (APC) to the TCR in the context of the appropriate major histocompatibility complex (MHC) class molecule (signal 1) and engagement of a co-stimulatory receptor (signal 2), the most important of which is mediated through the interaction of CD28 on T cells and CD80/CD86 on APC. Other cytokine signals from APC or regulatory T cells can amplify or diminish immune responses (signal 3). T-cell

Anti-CTLA4 clinical results

Two anti-CTLA-4 antibodies, ipilimumab and tremelimumab, have undergone extensive clinical evaluation. Ipilimumab is a fully human IgG1 monoclonal antibody that binds to the CTLA-4 receptor expressed on activated T cells. Phase I and II studies of ipilimumab established a biologically active and tolerable dose; although a new class of immune-related toxicities were observed (see below). These early studies also established that patients with advanced melanoma had more objective tumor

PD-1: Pre-clinical observations

The main pathway that negatively regulates the behavior of T cells in the periphery (e.g. at sites of chronic infection or tumors) is the programmed death-1 (PD-1) pathway. PD-1 is a transmembrane protein receptor up-regulated on T cells after TCR engagement. The ligands for this receptor, (PD-L1 (B7-H1) and PD-L2 (B7-DC)), found on APC, macrophages, T cells and B cells are up-regulated at the time of T-cell activation. PD-L1 is also expressed by many tumors including melanoma (Haile et al.,

Anti-PD-1 clinical results

The first anti-PD-1 antibody tested in patients with melanoma was MDX-1106, a fully human IgG4, now referred to as nivolumab (Brahmer et al., 2010). This antibody blocks the interaction between PD-L1 and PD-1 and also the interaction between PD-1 and CD80 found on B cells and macrophages whose normal function is to provide a costimulatory signal when it engages CD28 on activated T cells. A total of 39 patients participated in the phase I study, which also included other cancer types (advanced

Combination antibody therapy

As detailed above, monotherapy using antagonistic antibodies to CTLA-4, PD-1 or PD-L1 can induce significant tumor regressions and improve survival in patients with melanoma as well as other malignancies. There is also a strong pre-clinical rationale for blocking both CTLA-4 and PD-1 T-cell checkpoints in patients with melanoma. In a phase I study that investigated sequential and concurrent administration of ipilimumab and nivolumab (Wolchok et al., 2013). 17 of 53 patients received concurrent

Monoclonal antibodies under development for the treatment of melanoma and other solid tumors

Manipulation of other T-cell checkpoint and co-stimulatory pathways has high potential to improve melanoma therapy. Table 4 summarizes antibodies under development with application to melanoma.

These pathways are of interest not only for their ability to promote T cell anti-tumor responses, but also because they are present in the TIL in melanoma and other tumors. Gros et al. (2014) reported on the extensive characterization of the TIL from 6 patients with melanoma. The expression of multiple

OX40

OX40 is a member of the tumor necrosis factor receptor (TNFR) family that has a unique pattern of expression; it is for the most part restricted to lymphoid tissue and mainly expressed on activated CD4+ and CD8+ T cells (Baum et al., 1994, Paterson et al., 1987). Effector CD4+ T cells, which upregulate OX40 expression more rapidly than naïve T cells, express OX40 within 4 h after Ag stimulation (Gramaglia et al., 1998). OX40+ T cells are found preferentially at sites of inflammation and not

Anti-OX40 in patients with advanced cancer

A phase I trial tested three doses of a murine anti-human OX40 agonist antibody administered over five days (Curti et al., 2013). Thirty patients were treated with anti-OX40; ten at each dose level (0.1 mg/kg, 0.4 mg/kg and 2 mg/kg). Toxicities were mild; grade 1–2 fatigue and transient lymphopenia were the most common side effects observed. Toxicities generally resolved within 72 h of the last dose of anti-OX40. Immune-mediated diarrhea, colitis or endocrinopathies, which have been associated with

4-1BB

Another important T cell co-stimulatory pathway is 4-1BB (CD137), also part of the TNFR family. When activated, 4-1BB induces Bcl2 and BCL-xl resulting in decreased T-cell apoptosis (Hernandez-Chacon et al., 2011). There is also enhanced tumor-specific T-cell cytotoxicity and NK activity after 4-1BB agonist administration. Anti-tumor activity has been observed in several murine models mediated by CD8+ T cells, involves interferon-gamma secretion and results in long-term memory against tumor

Conclusions

Manipulation of the regulatory pathways that influence tumor-reactive T cells has led to FDA approval of several new antibody monotherapies for patients with advanced melanoma. The investigation of inhibitory (checkpoint) and co-stimulatory pathways that influence T cell anti-tumor activity for clinical benefit is just beginning and appears to apply to many different cancers. The early results from combination antibody therapy attempting to influence both the CTLA-4 and PD-1 pathways are even

References (75)

  • P.A. Ascierto et al.

    Clinical experience with ipilimumab 3 mg/kg: real-world efficacy and safety data from an expanded access programme cohort

    J. Transl. Med.

    (2014)
  • L. Baitsch et al.

    Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization

    PLoS ONE

    (2012)
  • D.L. Barber et al.

    Restoring function in exhausted CD8 T cells during chronic viral infection

    Nature

    (2006)
  • J. Baselga et al.

    Phase II multicenter study of the antiepidermal growth factor receptor monoclonal antibody cetuximab in combination with platinum-based chemotherapy in patients with platinum-refractory metastatic and/or recurrent squamous cell carcinoma of the head and neck

    J. Clin. Oncol.

    (2005)
  • P.R. Baum et al.

    Molecular characterization of murine and human OX40/OX40 ligand systems: identification of a human OX40 ligand as the HTLV-1-regulated protein gp34

    EMBO J.

    (1994)
  • J.A. Bluestone et al.

    Immunosuppressive effects of anti-CD3 MAb and soluble co-stimulatory molecules

    Transplant. Proc.

    (1993)
  • J.R. Brahmer et al.

    Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates

    J. Clin. Oncol.

    (2010)
  • J.R. Brahmer et al.

    Safety and activity of anti-PD-L1 antibody in patients with advanced cancer

    N. Engl. J. Med.

    (2012)
  • A.C. Buenafe et al.

    V beta CDR3 motifs associated with BP recognition are enriched in OX-40+ spinal cord T cells of Lewis rats with EAE

    J. Neurosci. Res.

    (1996)
  • D. Cunningham et al.

    Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer

    N. Engl. J. Med.

    (2004)
  • M.A. Curran et al.

    PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors

    Proc. Natl. Acad. Sci. U.S.A.

    (2010)
  • B.D. Curti et al.

    OX40 is a potent immune-stimulating target in late-stage cancer patients

    Cancer Res.

    (2013)
  • T.A. Davis et al.

    Rituximab anti-CD20 monoclonal antibody therapy in non-Hodgkin's lymphoma: safety and efficacy of re-treatment

    J. Clin. Oncol.

    (2000)
  • H. Dong et al.

    Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion

    Nat. Med.

    (2002)
  • A.M. Eggermont et al.

    Ipilimumab versus placebo after complete resection of stage III melanoma: initial efficacy and safety results from the EORTC 18071 phase III trial

    J. Clin. Oncol.

    (2014)
  • L.A. Fecher et al.

    Ipilimumab and its toxicities: a multidisciplinary approach

    Oncologist

    (2013)
  • R.L. Ferris et al.

    Tumor antigen-targeted, monoclonal antibody-based immunotherapy: clinical response, cellular immunity, and immunoescape

    J. Clin. Oncol.

    (2010)
  • L.M. Francisco et al.

    PD-L1 regulates the development, maintenance, and function of induced regulatory T cells

    J. Exp. Med.

    (2009)
  • D.T. Frederick et al.

    BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma

    Clin. Cancer Res.

    (2013)
  • G.J. Freeman

    Structures of PD-1 with its ligands: sideways and dancing cheek to cheek

    Proc. Natl. Acad. Sci. U.S.A.

    (2008)
  • I. Gramaglia et al.

    Ox-40 ligand: a potent costimulatory molecule for sustaining primary CD4 T cell responses

    J. Immunol.

    (1998)
  • A. Gros et al.

    PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors

    J. Clin. Invest.

    (2014)
  • S.T. Haile et al.

    Tumor cell programmed death ligand 1-mediated T cell suppression is overcome by coexpression of CD80

    J. Immunol.

    (2011)
  • O. Hamid et al.

    Clinical activity, safety and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM)

    J. Clin. Oncol.

    (2013)
  • R.S. Herbst et al.

    Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients

    Nature

    (2014)
  • J.A. Hernandez-Chacon et al.

    Costimulation through the CD137/4-1BB pathway protects human melanoma tumor-infiltrating lymphocytes from activation-induced cell death and enhances antitumor effector function

    J. Immunother.

    (2011)
  • F.S. Hodi et al.

    Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • Cited by (0)

    This article belongs to Special Issue on Therapeutic Antibodies.

    View full text