Open access peer-reviewed chapter

Immune Checkpoint Blockade and Adaptive Immune Resistance in Cancer

Written By

Raymond M. Wong and Robert B. Cameron

Submitted: 02 June 2016 Reviewed: 24 October 2016 Published: 26 April 2017

DOI: 10.5772/66494

From the Edited Volume

Immunotherapy - Myths, Reality, Ideas, Future

Edited by Krassimir Metodiev

Chapter metrics overview

1,858 Chapter Downloads

View Full Metrics

Abstract

The clinical success of immune checkpoint blockers is a pivotal advancement for treating an increasing number of cancer types. However, immune checkpoint blockers still rarely induce complete remission and show little to no therapeutic efficacy in a significant percentage of cancer patients. Efforts are now underway to identify biomarkers that accurately predict which patients benefit from immune checkpoint blockers. Moreover, adaptive immune resistance can develop in tumors during treatment with immune checkpoint blockers. These adaptive resistance mechanisms in tumors might be disrupted by combining adjunctive immunotherapies, which could potentially improve the therapeutic efficacy of immune checkpoint blockers. This chapter discusses the mechanism of action of cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint blockers and biomarkers that might predict clinical responses to these drugs. Lastly, ongoing research on mechanisms of tumor adaptive resistance could facilitate rationale design of adjunctive immunotherapies that can be synergistically combined with immune checkpoint blockers to more effectively treat cancer.

Keywords

  • immunotherapy
  • T lymphocytes
  • immune checkpoints
  • CTLA-4
  • PD-1
  • PD-L1

1. Introduction

Immune checkpoints are inhibitory pathways that are critical for maintaining self-tolerance. Immune checkpoints also control the magnitude and duration of physiological immune responses in peripheral tissues in order to minimize collateral damage. Immune checkpoint receptors and their cognate ligands are naturally expressed on a variety of cell types, including antigen-presenting cells, T cells, B cells, tumor cells, tumor stroma, and also normal tissue. A number of immune checkpoint pathways have been identified, including cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed death-1 (PD-1), programmed death ligand-1 (PD-L1), T cell immunoglobulin and mucin domain 1 (TIM-1), T cell immunoglobulin and mucin domain 3 (TIM-3), lymphocyte-activation gene 3 (LAG-3), T cell immunoreceptor with Ig and ITIM domains (TIGIT), V-domain Ig suppressor of T cell activation (VISTA), carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1), herpesvirus entry mediator (HVEM), B- and T-lymphocyte attenuator (BTLA), CD160, CD200, CD200 receptor, and adenosine 2A receptor (A2Ar). For brevity, this chapter will focus on CTLA-4 and PD-1/PD-L1, as clinical drugs targeting these pathways have been successfully developed to treat an increasing variety of human cancer types.

Advertisement

2. Main body

2.1. CTLA-4

CTLA-4 is the first immune checkpoint receptor to be clinically targeted. CTLA-4 is expressed mainly on the surface of activated T cells. While certain subsets of T regulatory cells constitutively express CTLA-4, it is virtually undetectable on naïve, inactivated T cells. Upon activation, both CD4+ and CD8+ T cells upregulate CTLA-4 on the surface, reaching maximum level within 2–3 days. CD4+ T cells are reported to express more CTLA-4 mRNA and protein compared to CD8+ T cells, suggesting that CTLA-4 has a more significant regulatory effect on CD4+ T cells [1].

CTLA-4 downregulates T cell activation by sequestering CD80 and CD86 costimulatory molecules on antigen-presenting cells. This prevents CD80 and CD86 from delivering costimulatory activation signals to T cells through the CD28 receptor. CTLA-4 binds to CD80 and CD86 with ~10 times higher affinity than CD28 [2]. CTLA-4 expressed on T cells can also remove CD80 and CD86 molecules from neighboring antigen-presenting cells through a process called trans-endocytosis [3]. CTLA-4 also prevents CD28 recruitment to the immunological synapse, further impairing T cell activation [4].

CTLA-4 knockout mice die within 2–3 weeks of age due to massive lymphoproliferation, resulting in destruction of vital organs [5]. This lethal phenotype is associated primarily with hyperactivated CD4+ T cells, which are skewed toward a T helper type-2 phenotype and have increased resistance to apoptosis. These hyperactivated CD4+ T cells abnormally infiltrate into peripheral tissues, resulting in organ failure. These observations led cancer immunology researchers to hypothesize that blockade of CTLA-4 signaling could potentially induce effective T cell-mediated immune responses against tumor tissue.

A pivotal laboratory study reported in 1996 by James Allison’s group showed that treatment of tumor-bearing mice with a CTLA-4-blocking antibody could effectively induce tumor regression [6]. Despite much subsequent investigation, the in vivo mechanism of action of CTLA-4 blockade immunotherapy has remained elusive. The prevailing hypothesis is that CTLA-4 blockade not only enhances T cell infiltration into tumors but also reduces the relative presence of immunosuppressive T regulatory cells in tumor tissue [7]. This alteration in the ratio of effector T cells versus T regulatory cells in tumors tilts the immunological balance in favor of T cell-mediated destruction of tumor cells.

These studies led to pharmaceutical development of the first immune checkpoint blocker, ipilimumab (Yervoy®). Ipilimumab is a fully human monoclonal antibody that blocks the CTLA-4 receptor, thereby preventing its ability to sequester CD80 and CD86 costimulatory molecules. It was initially tested in melanoma, and demonstrated extended overall survival in patients versus a comparator melanoma peptide-based immunotherapy vaccine called gp100. In a randomized phase III clinical trial, melanoma patients receiving ipilimumab had a median overall survival of 10.4 months versus 6.4 months in those receiving only the gp100 peptide vaccine (Hodi 2010). Objective response rates (measureable tumor regression) were 10.9% in the ipilimumab group versus 1.5% in the gp100 vaccine group. The responses to ipilimumab were durable, with the 1-year and 2-year survival rate being 46 and 24%, respectively. By comparison, the 1-year and 2-year survival rate in patients receiving only the gp100 peptide vaccine was only 25 and 14%, respectively [8]. These trial results led to US FDA approval of ipilimumab for melanoma in 2011.

2.2. PD-1

PD-1 is another major immune checkpoint receptor that regulates T cell activity against tumor tissue. PD-1 is a cell surface receptor originally identified in a murine T cell hybridoma undergoing programmed cell death [9]. PD-1 is absent on naïve inactivated immune cells but is significantly upregulated on activated T cells, B cells, natural killer cells and myeloid-derived cells [10]. In T cells, PD-1 expression is induced by T cell receptor signaling [11] and also by certain pro-inflammatory cytokines including interleukin-2, interleukin-7, interleukin-15, and interleukin-21 [12].

PD-1 signaling downregulates T cell activity primarily via interaction with its two natural ligands: Programmed Death Ligand-1 (PD-L1) and Programmed Death Ligand-2 (PD-L2). PD-L1 is expressed on a wide variety of cell types including hematopoietic cells, T cells, B cells, myeloid cells, and dendritic cells [10]. It is also expressed on a wide variety of peripheral tissues such as skeletal muscle, lung, heart, and placenta [10]. Notably, PD-L1 is also expressed on a wide variety of cancer cells and generally is associated with poorer patient prognosis [13]. PD-L2 expression is generally more restricted, being found primarily on dendritic cells, macrophages, and occasionally cancer cells [14]. PD-L2 binds to PD-1 with two- to sixfold higher relative affinity than PD-L1 [15]. However, PD-L2 is generally expressed at lower relative levels [16]. Thus, it is believed that PD-L1 is the predominant ligand for PD-1.

Signaling through the PD-1 receptor on T cells results in downstream inhibition of PI3K/AKT activation [17]. The net effect is downregulation of a number of effector functions including cytokine secretion and cytolytic activity. PD-1 knockout mice have various autoimmune pathologies, including autoantibody-induced cardiomyopathy [18], arthritis and lupus-like disease [19], and diabetes [20]. In peripheral tissues, the immunosuppressive activity of PD-1 is mediated primarily by interaction with PD-L1 [21]. PD-L1 expressed in tumor tissue also impairs host antitumor immune responses [22]. PD-L1 and/or PD-L2 in tumor tissue facilitates evasion from host immune responses via multiple mechanisms including induction of T cell anergy and exhaustion [23], promoting T cell apoptosis [24], and also by enhancing the expansion and activity of immunosuppressive T regulatory cells [25]. Moreover, PD-1 can transmit an antiapoptotic signal to PD-L1-expressing tumor cells, which renders them resistant to lysis by cytotoxic T lymphocytes [26].

This fundamental understanding of the PD-1/PD-L1 axis in suppressing host antitumor immune responses led to development of the first clinical PD-1 blockers, nivolumab (Opdivo®) and pembrolizumab (Keytruda®). Both nivolumab and pembrolizumab are fully human monoclonal antibodies that block the PD-1 receptor, thereby preventing its ability to bind its natural ligands PD-L1 and PD-L2. In large phase I clinical trials, nivolumab and pembrolizumab each demonstrated durable clinical response rates with acceptable safety profiles in patients with advanced melanoma, non-small cell lung cancer, renal cell carcinoma or Hodgkin’s lymphoma [2730]. Nivolumab and pembrolizumab are now both FDA approved for treating melanoma and non-small cell lung cancer. Nivolumab is additionally approved for treating renal cell carcinoma, Hodgkin’s lymphoma, and also for use in combination with the CTLA-4 blocker, ipilimumab, for treating melanoma. Remarkably, in two separate melanoma clinical trials, the combination of nivolumab and ipilimumab induced objective responses in ~60% of patients, with complete responses seen in ~11.5–22% of patients [3132].

Pembrolizumab and nivolumab (and a third investigational PD-1 blocker, pidilizumab) are now collectively continuing in 500+ clinical trials. Virtually all cancer types are now being targeted with PD-1/PD-L1 blockers in some capacity. Notably, there is a significant effort to test nivolumab or pembrolizumab with other adjunctive therapies to determine synergistic combinatorial regimens. Conventional treatments like chemotherapy and radiation have shown in animal tumor models to potentially synergize with PD-1/PD-L1 blockers [3335]. In addition, PD-1 blockers are now also being tested in combination with small molecule drugs (investigational and Food and Drug Administration (FDA) approved) and also experimental immunotherapies such as vaccines and chimeric antigen receptor T cells.

All clinical PD-1 blockers have the same mechanism of action. Slight variances in the protein structure among different PD-1 blockers could potentially confer differences in binding affinity for the PD-1 receptor and also differences in half-life (i.e. persistence in the body). The physiological significance and clinical effectiveness of such variances remain undetermined.

2.3. PD-L1

Expression of PD-L1 is found on diverse cell types, including normal and malignant tissue, antigen presenting cells, myeloid cells, B cells, and T cells. PD-L1 downregulates T cells via multiple mechanisms. PD-L1 expressed on various cells primarily interacts with PD-1 expressed on T cells, delivering an inhibitory signal that downregulates T cell activity. PD-L1 also binds to CD80 expressed on both antigen-presenting cells and activated T cells [36]. Interaction of PD-L1 with CD80 on antigen-presenting cells prevents CD80 from delivering costimulatory activating signals to T cells. When PD-L1 binds to CD80 expressed on activated T cells, an inhibitory signal is delivered to T cells. Currently, it is unknown exactly what intracellular signaling pathways are altered when PD-L1 binds to CD80 on T cells. Nonetheless, it is now generally understood that blocking PD-L1 results in enhanced T cell activation.

Atezolizumab (Tecentriq®) was the first PD-L1 blocker to enter clinical trials. Atezolizumab is a fully human monoclonal antibody that prevents PD-L1 from binding to PD-1 and CD80. It was initially tested in patients with PD-L1-positive metastatic bladder cancer [37]. Bladder cancer patients with PD-L1-negative tumors were subsequently included for treatment. Clinical response rates were ~15% of PD-L1-negative patients and ~25% of PD-L1-positive patients [37]. Because of the higher clinical activity of atezolizumab in PD-L1-positive bladder cancer, a companion diagnostic called the Ventana PD-L1 (SP142) assay is offered to provide tumor PD-L1 expression status of patients considering atezolizumab treatment. In 2016, atezolizumab was FDA approved for urothelial carcinoma, the most common form of bladder cancer. Like nivolumab and pembrolizumab PD-1 blockers, atezolizumab is now continuing in clinical trials for a wide variety cancer types and also being tested in combination with conventional cancer treatments, small molecule drugs and other investigational immunotherapies. Alternative PD-L1 blockers, such as avelumab and durvalumab, are also now in clinical trials.

2.4. Predictive biomarkers for CTLA-4 and PD-1/PD-L1 blockers

CTLA-4 and PD-1/PD-L1 immune checkpoint blockers have proven to be pivotal advancements in cancer treatment. However, a significant proportion of cancer patients still experience little to no clinical benefit from treatment. Even among responding patients, only a small minority achieve complete remission. Studies using clinical tumor specimens from patients treated with immune checkpoint blockers have revealed some potentially important differences between responders versus nonresponders.

During early clinical development of PD-1 blockers, it was hypothesized that differential expression levels of PD-L1 in tumor tissue would correlate with clinical responses. It was anticipated that PD-L1 expression in tumor tissue could therefore be a predictive biomarker to accurately identify patients likely to respond to PD-1 or PD-L1 blockers. However, a definitive correlation has thus far not been established. Both PD-L1-positive and PD-L1-negative tumors can respond to PD-1 or PD-L1 blockers. Further confounding factors include variability of PD-L1 expression in different anatomical areas of tumor tissue. In addition, PD-L1 expression in tumor tissue may be transient—appearing and disappearing due to treatments or other poorly understood influences. Lastly, assays measuring PD-L1 in tumors have yet to establish a clear threshold of expression that defines what is considered “PD-L1-positive.” For instance, the FDA-approved Ventana PD-L1 assay defines ≥5% PD-L1-positive cells in bladder cancer tissue to be associated with higher clinical response rates to atezolizumab [38]. However, alternative PD-L1 assays used in various other clinical trials of nivolumab or pembrolizumab have wide variability in PD-L1 expression analysis methodologies. Overall, it is generally agreed upon that low or absent PD-L1 expression in tumors is not sufficient to preclude a patient from treatment with PD-1/PD-L1 blockers [39].

Alternative predictive biomarkers for clinical response to PD-1/PD-L1 blockers are currently being explored. CD8+ T cell infiltration into tumors might be predictive of clinical response to PD-1 blockers. Specifically, the density of pretreatment CD8+ T cells at both the tumor invasive margin and tumor center may be correlated with clinical response to pembrolizumab. In serially biopsied tumors from melanoma patients undergoing pembrolizumab treatment, it was shown that responding patients generally had higher densities of CD8+/PD-1+ cells in close proximity to PD-L1-expressing tumor cells [40]. Furthermore, serial analysis of tumor biopsies showed that intratumoral CD8+/PD-1+ T cells actively proliferate during pembrolizumab treatment [40]. These data offer insights on a potential mechanism of PD-1 blockade efficacy, whereby presence of pretreatment CD8+ T cells in tumors is a prerequisite for clinical response. However, like tumor PD-L1 expression assays, establishing a standard cut-off threshold value for CD8+ T cell levels in tumors that accurately predicts clinical response to PD-1/PD-L1 blockade will be challenging. Tumors of various tissue origins often contain infiltrating T cells that can vary greatly in absolute number, density, and also anatomical location within the intratumoral space. Nonetheless, establishing a “scoring system” based on pretreatment CD8+ T cell infiltration warrants further investigation as a potential predictive biomarker.

Another intriguing biomarker with predictive potential may be intratumoral expression of indoleamine-2,3-dioxygenase (IDO). IDO is a tryptophan catabolizing enzyme that is occasionally expressed in various tumor types. Depletion of tryptophan within tumors by IDO may be a rate-limiting step for effective antitumor T cell activity. Studies in melanoma patients treated with ipilimumab suggest a correlation between pretreatment IDO expression and clinical response. In one study, intratumoral IDO was detected in 37.5% of responding melanoma patients and only 11.1% in nonresponders [41]. It remains to be seen if similar patterns are seen in other cancer types and also patients treated with PD-1/PD-L1 blockers.

Genetic signatures of tumors are yet another parameter with potential for yielding predictive biomarkers for clinical response to immune checkpoint blockers. Certain tumors, such as colorectal cancer, are highly refractory to treatment with PD-1 blockers. In early clinical trials of nivolumab, it was found that only 1 in 33 colorectal cancer patients responded to treatment [2728]. Subsequently, it was hypothesized that the single responding colorectal cancer patient harbored a defect in DNA mismatch repair in tumor tissue, resulting in a significantly high load of somatic mutations [42]. Defects in tumor tissue mismatch repair can result in thousands of somatic mutations, providing a larger pool of neo-antigens for immune recognition. Immune checkpoint blockade therapy could therefore amplify the natural adaptive immune response to mutated neo-antigens. Hence, mutational load in pretreatment tumor tissue might be predictive of clinical response to immune checkpoint blockers. To test this hypothesis, a small clinical trial focusing primarily on colorectal cancer showed that patients with defects in tumor tissue mismatch repair harbored significantly higher loads of somatic mutations versus those with mismatch repair-proficient tumors. Upon treatment with pembrolizumab, higher response rates and longer survival times were seen in patients with mismatch repair defects versus those with proficient mismatch repair [42]. This pivotal study has catalyzed further investigation of tumor mutational profiles to determine if a correlation with clinical responses can be established in large studies of diverse cancer types.

2.5. Adaptive immune resistance

Mechanisms of inherent and acquired resistance to immune checkpoint blockade are poorly understood. Clinical responses to CTLA-4 and PD-1/PD-L1 blockers are often durable, sometimes lasting years. However, complete regressions are still relatively rare and eventual disease relapse among responding patients is frequent. Recent studies have offered insights that immunological parameters of tumor tissue adapt in response to T cell-mediated attack induced by immune checkpoint blockers. Enhanced T cell activity within tumors involves local production of inflammatory mediators, such as interferon (IFN)-γ, which is known to upregulate PD-L1 on peripheral tissues [43]. Upregulation of PD-L1 on various cell types within tumor tissue might result in heightened CD80-mediated inhibition of proximal effector T cells.

Furthermore, augmentation of effector T cell activity in tumor tissue via PD-1 blockade may subsequently induce compensatory upregulation of alternative immune checkpoint receptors, TIM-3. TIM-3 is a receptor expressed primarily on IFN-γ-secreting CD4+ and CD8+ T cells [44]. TIM-3 is bound by multiple ligands, including galectin-9, CEACAM-1, and high-mobility group box 1 (HMGB-1). Signaling through TIM-3 in activated T cells triggers the release of human leukocyte antigen B-associated transcript 3 (BAT3) from the TIM-3 cytoplasmic domain. This results in defective production of IL-2, IFN-γ, and likely other pro-inflammatory cytokines [44]. Although the TIM-3 signaling pathway has yet to be fully elucidated, it seems clear that TIM-3 affects T cell receptor downstream signaling via a mechanism distinct from PD-1 and CTLA-4.

TIM-3 appears to be co-expressed with PD-1 in tumor-infiltrating lymphocytes of cancer patients and is upregulated on T cells upon therapeutic PD-1 blockade [45]. This may provide a mechanism of immunological escape and a possible reason for incomplete clinical responses upon PD-1 blockade immunotherapy. It might also be a contributing factor toward acquired resistance to PD-1 blockade clinically, whereby patients initially respond to treatment but eventually relapse despite continuous therapy. Preclinical studies in animal tumor models show that PD-1 blockade immunotherapy results in upregulation of TIM-3 on T cells. Co-blockade of both TIM3 and PD-1 can prevent resistance to PD-1 blockade immunotherapy [45]. As such, TIM-3 blocking antibodies are now in early phase clinical trials to evaluate their safety, tolerability, and dosing ranges. Figure 1 illustrates how PD-1/PD-L1 blockade may result in compensatory upregulation of TIM-3 and/or PD-L1 on T cells and tumor cells.

Figure  1.

PD-1/PD-L1 blockade promotes T cell-mediated inflammation in tumors. In turn, this can trigger upregulation of PD-L1 on various cells within tumor tissue. This can also trigger compensatory upregulation of TIM-3 on effector T cells. Upregulation of PD-L1 and TIM-3, even during continuous treatment with PD-1 blockers, can impair T cell activity and result in clinical resistance.

Downregulation of major histocompatibility (MHC) receptor expression in tumors might also contribute to acquired resistance to PD-1 blockers. Loss-of-function mutations in the MHC beta-2 microglobulin antigen-presenting protein have been noted in selected melanoma patients who initially responded to pembrolizumab therapy but subsequently relapsed [46]. Further studies in larger patient populations are necessary to confirm the association of MHC-related mutations and acquired resistance to PD-1 blockers.

2.6. Strategies to counteract adaptive resistance to immune checkpoint blockade

The mechanism of inherent and acquired/adaptive resistance to CTLA-4 and PD-1/PD-L1 immune checkpoint blockers is not fully understood and could possibly vary between individual patients and different tumor types. However, research on predictive biomarkers and mechanisms of adaptive resistance to PD-1 blockers have yielded insight that might be extrapolated to rationally design combination immunotherapies that synergistically enhance the efficacy of immune checkpoint blockers. For instance, it is now generally understood that PD-1 blockers augment T cell-mediated inflammation in tumor tissue. In turn, this can promote upregulation of PD-L1 on various cells in tumors, likely due to IFN-γ signaling [43]. Upregulation of PD-L1 expression in tumor tissue can promote enhanced CD80 signaling in T cells, which impairs T cell activity [36]. PD-1 blockade may also induce compensatory upregulation of alternative immune checkpoint receptors, such as TIM-3, on T cells within tumor tissue [45]. TIM-3 signaling results in downregulation of T cell activity. Next-generation immunotherapeutic regimens might combine PD-1 blockers such as nivolumab/pembrolizumab with PD-L1 blockers like atezolizumab, to counteract PD-L1 upregulation induced by T cell-mediated inflammation in tumor tissue. Other rational combinations might include PD-1/PD-L1 blockers combined with investigational TIM-3 blockers, to counteract the effects of TIM-3 upregulation on activated T cells.

Another strategy to enhance the efficacy of immune checkpoint blockers might involve improving T cell trafficking to tumor tissue. The extent of T cell infiltration into tumor tissue may be a predictive biomarker and a prerequisite for efficacy of both CTLA-4 and PD-1/PD-L1 blockers. As such, therapies that promote T cell trafficking to tumors could potentially improve tumor sensitivity to immune checkpoint blockers. Studies of human melanoma tumors have identified a set of chemokines that are associated with enhanced recruitment of T cells toward tumor tissue. These chemokines, including CCL2, CCL3, CCL4, CCL5, CXCL9, and CXCL10, might have utility as clinical therapies to improve T cell trafficking to tumors [47]. However, such chemokines or other T cell recruitment factors must be targeted specifically to tumor tissue in order to effectively recruit T cells. T cell recruitment factors might be coupled to antibodies that bind to tumor cell receptors, thus providing a vehicle for tumor targeting. In animal tumor studies, a T cell recruitment factor called LIGHT (also called tumor necrosis factor superfamily member 14) was fused to an anti-epidermal growth factor receptor (EGFR) antibody. This LIGHT-anti-EGFR fusion molecule was able to promote more extensive T cell infiltration into EGFR-expressing tumors. In turn, this prevented resistance to PD-L1 blockade immunotherapy [48]. Similar strategies that target other T cell recruitment factors toward tumors might be feasible.

Our group at the Pacific Heart, Lung & Blood Institute (Los Angeles, CA) is conducting research on gene-modified human mesenchymal stem cells (MSCs) as a strategy to alter the tumor microenvironment and prevent resistance to immune checkpoint blockers. MSCs can be isolated and expanded from various adult tissues including bone marrow, fat, umbilical cord blood, and term placentas. MSCs are known to preferentially migrate to tumor tissue, making them potentially useful drug delivery vectors to alter the immunological microenvironment of tumors [49]. In animal tumor models, MSCs have been genetically modified in diverse ways to effectively treat tumors. These include modification to produce immunostimulatory cytokines (e.g. IFN-α, IFN-β, IL-12) and T cell trafficking molecules such as LIGHT [5053].

Both autologous and allogeneic MSCs have been used extensively in clinical trials for treating severe inflammatory disorders and certain degenerative conditions, and generally have an acceptable safety profile [54]. Autologous gene-modified MSCs have recently entered clinical trials for cancer [55]. It remains to be seen if MSCs and other tumor-targeting systems can effectively deliver pro-inflammatory agents to tumor tissue and improve sensitivity to clinical immune checkpoint blockers.

Advertisement

Acknowledgments

Research funding at the Pacific Heart, Lung & Blood Institute is provided in part by grants from the Richard M. Schulze Family Foundation, the H.N. & Frances C. Berger Foundation, and the Kazan McClain Partners’ Foundation.

References

  1. 1. Chan DV, Gibson HM, Aufiero BM, Wilson AJ, Hafner MS, Mi QS, Wong HK. Differential CTLA-4 expression in human CD4+ versus CD8+ T cells is associated with increased NFAT1 and inhibition of CD4+ proliferation. Genes Immun. 15(1):25–32; 2014.
  2. 2. van der Merwe PA, Bodian DL, Daenke S, Linsley P, Davis SJ. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J Exp Med. 185(3):393–403; 1997.
  3. 3. Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM, Baker J, Jeffery LE, Kaur S, Briggs Z, Hou TZ, Futter CE, Anderson G, Walker LS, Sansom DM. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science. 332(6029):600–3; 2011.
  4. 4. Greene JL, Leytze GM, Emswiler J, Peach R, Bajorath J, Cosand W, Linsley PS. Covalent dimerization of CD28/CTLA-4 and oligomerization of CD80/CD86 regulate T cell costimulatory interactions. J Biol Chem. 271(43):26762–71; 1996.
  5. 5. Khattri R, Auger JA, Griffin MD, Sharpe AH, Bluestone JA. Lymphoproliferative disorder in CTLA-4 knockout mice is characterized by CD28-regulated activation of Th2 responses. J Immunol. 162(10):5784–91; 1999.
  6. 6. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 271(5256):1734–6; 1996.
  7. 7. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 161(2):205–14; 2015.
  8. 8. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 363(8):711–23; 2010.
  9. 9. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 11(11):3887–95; 1992.
  10. 10. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 26:677–704; 2008.
  11. 11. Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 8(5):765–72; 1996.
  12. 12. Kinter AL, Godbout EJ, McNally JP, Sereti I, Roby GA, O'Shea MA, Fauci AS. The common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J Immunol. 181(10):6738–46; 2008.
  13. 13. Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med. 21(1):24–33; 2015.
  14. 14. Rozali EN, Hato SV, Robinson BW, Lake RA, Lesterhuis WJ. Programmed death ligand 2 in cancer-induced immune suppression. Clin Dev Immunol. 2012:656340; 2012.
  15. 15. Youngnak P, Kozono Y, Kozono H, Iwai H, Otsuki N, Jin H, Omura K, Yagita H, Pardoll DM, Chen L, Azuma M. Differential binding properties of B7-H1 and B7-DC to programmed death-1. Biochem Biophys Res Commun. 307(3):672–7; 2003.
  16. 16. Liang SC, Latchman YE, Buhlmann JE, Tomczak MF, Horwitz BH, Freeman GJ, Sharpe AH. Regulation of PD-1, PD-L1, and PD-L2 expression during normal and autoimmune responses. Eur J Immunol. 33(10):2706–16; 2003.
  17. 17. Riley JL. PD-1 signaling in primary T cells. Immunol Rev. 229(1):114–25; 2009.
  18. 18. Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, Sasayama S, Mizoguchi A, Hiai H, Minato N, Honjo T. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 291(5502):319–22; 2001.
  19. 19. Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 11(2):141–51; 1999.
  20. 20. Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A. 102(33):11823–8; 2005.
  21. 21. Tsushima F, Yao S, Shin T, Flies A, Flies S, Xu H, Tamada K, Pardoll DM, Chen L. Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood. 110(1):180–5; 2007.
  22. 22. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 8(6):467–77; 2008.
  23. 23. Crespo J, Sun H, Welling TH, Tian Z, Zou W. T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr Opin Immunol. 25(2):214–21; 2013.
  24. 24. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 8(8):793–800; 2002.
  25. 25. Amarnath S, Mangus CW, Wang JC, Wei F, He A, Kapoor V, Foley JE, Massey PR, Felizardo TC, Riley JL, Levine BL, June CH, Medin JA, Fowler DH. The PDL1-PD1 axis converts human TH1 cells into regulatory T cells. Sci Transl Med. 3(111):111ra120; 2011.
  26. 26. Azuma T, Yao S, Zhu G, Flies AS, Flies SJ, Chen L. B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood. 111(7):3635–43; 2008.
  27. 27. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, Gilson MM, Wang C, Selby M, Taube JM, Anders R, Chen L, Korman AJ, Pardoll DM, Lowy I, Topalian SL. 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. 28(19):3167–75; 2010.
  28. 28. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 366(26):2443–54; 2012.
  29. 29. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, Rodig SJ, Chapuy B, Ligon AH, Zhu L, Grosso JF, Kim SY, Timmerman JM, Shipp MA, Armand P. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 372(4):311–9; 2015.
  30. 30. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, Dronca R, Gangadhar TC, Patnaik A, Zarour H, Joshua AM, Gergich K, Elassaiss-Schaap J, Algazi A, Mateus C, Boasberg P, Tumeh PC, Chmielowski B, Ebbinghaus SW, Li XN, Kang SP, Ribas A. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 369(2):134–44; 2013.
  31. 31. Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, Linette GP, Meyer N, Giguere JK, Agarwala SS, Shaheen M, Ernstoff MS, Minor D, Salama AK, Taylor M, Ott PA, Rollin LM, Horak C, Gagnier P, Wolchok JD, Hodi FS. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 372(21):2006–17; 2015.
  32. 32. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K, Burke MM, Caldwell A, Kronenberg SA, Agunwamba BU, Zhang X, Lowy I, Inzunza HD, Feely W, Horak CE, Hong Q, Korman AJ, Wigginton JM, Gupta A, Sznol M. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 369(2):122–33; 2013.
  33. 33. Rios-Doria J, Durham N, Wetzel L, Rothstein R, Chesebrough J, Holoweckyj N, Zhao W, Leow CC, Hollingsworth R. Doxil synergizes with cancer immunotherapies to enhance antitumor responses in syngeneic mouse models. Neoplasia. 17(8):661–70; 2015.
  34. 34. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, Fu YX. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 124(2):687–95; 2014.
  35. 35. Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, Durham N, Meyer C, Harris TJ, Albesiano E, Pradilla G, Ford E, Wong J, Hammers HJ, Mathios D, Tyler B, Brem H, Tran PT, Pardoll D, Drake CG, Lim M. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. 86(2):343–9; 2013.
  36. 36. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 27(1):111–22; 2007.
  37. 37. Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, Bellmunt J, Burris HA, Petrylak DP, Teng SL, Shen X, Boyd Z, Hegde PS, Chen DS, Vogelzang NJ. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 515(7528):558–62; 2014.
  38. 38. McDermott DF, Sosman JA, Sznol M, Massard C, Gordon MS, Hamid O, Powderly JD, Infante JR, Fassò M, Wang YV, Zou W, Hegde PS, Fine GD, Powles T. Atezolizumab, an anti-programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: long-term safety, clinical activity, and immune correlates from a phase Ia study. J Clin Oncol. 34(8):833–42; 2016.
  39. 39. Grigg C, Rizvi NA. PD-L1 biomarker testing for non-small cell lung cancer: truth or fiction? J Immunother Cancer. 4:48; 2016.
  40. 40. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, West AN, Carmona M, Kivork C, Seja E, Cherry G, Gutierrez AJ, Grogan TR, Mateus C, Tomasic G, Glaspy JA, Emerson RO, Robins H, Pierce RH, Elashoff DA, Robert C, Ribas A. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 515(7528):568–71; 2014.
  41. 41. Hamid O, Schmidt H, Nissan A, Ridolfi L, Aamdal S, Hansson J, Guida M, Hyams DM, Gómez H, Bastholt L, Chasalow SD, Berman D. A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J Transl Med. 9:204; 2011.
  42. 42. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA Jr. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 372(26):2509–20; 2015.
  43. 43. Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, Chen S, Klein AP, Pardoll DM, Topalian SL, Chen L. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 4(127):127ra37; 2012.
  44. 44. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity. 44(5):989–1004; 2016.
  45. 45. Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG, Gandhi L, Redig AJ, Rodig SJ, Asahina H, Jones RE, Kulkarni MM, Kuraguchi M, Palakurthi S, Fecci PE, Johnson BE, Janne PA, Engelman JA, Gangadharan SP, Costa DB, Freeman GJ, Bueno R, Hodi FS, Dranoff G, Wong KK, Hammerman PS. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 7:10501; 2016.
  46. 46. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, Torrejon DY, Abril-Rodriguez G, Sandoval S, Barthly L, Saco J, Homet Moreno B, Mezzadra R, Chmielowski B, Ruchalski K, Shintaku IP, Sanchez PJ, Puig-Saus C, Cherry G, Seja E, Kong X, Pang J, Berent-Maoz B, Comin-Anduix B, Graeber TG, Tumeh PC, Schumacher TN, Lo RS, Ribas A. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 375(9):819–29; 2016.
  47. 47. Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 69(7):3077–85; 2009.
  48. 48. Tang H, Wang Y, Chlewicki LK, Zhang Y, Guo J, Liang W, Wang J, Wang X, Fu YX. Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell. 29(3):285–96; 2016.
  49. 49. Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev. 62(12):1156–66; 2010.
  50. 50. Studeny M, Marini FC, Dembinski JL, Zompetta C, Cabreira-Hansen M, Bekele BN, Champlin RE, Andreeff M. Mesenchymal stem cells: potential precursors for tumor stroma and targeted-delivery vehicles for anticancer agents. J Natl Cancer Inst. 96(21):1593–603; 2004.
  51. 51. Sartoris S, Mazzocco M, Tinelli M, Martini M, Mosna F, Lisi V, Indraccolo S, Moserle L, Cestari T, Riviera AP, Bifari F, Tridente G, Pizzolo G, Krampera M. Efficacy assessment of interferon-alpha-engineered mesenchymal stromal cells in a mouse plasmacytoma model. Stem Cells Dev. 20(4):709–19; 2011.
  52. 52. Jeong KY, Lee EJ, Kim SJ, Yang SH, Sung YC, Seong J. Irradiation-induced localization of IL-12-expressing mesenchymal stem cells to enhance the curative effect in murine metastatic hepatoma. Int J Cancer. 137(3):721–30; 2015.
  53. 53. Zou W, Zheng H, He TC, Chang J, Fu YX, Fan W. LIGHT delivery to tumors by mesenchymal stem cells mobilizes an effective antitumor immune response. Cancer Res. 72(12):2980–9; 2012.
  54. 54. Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, Granton J, Stewart DJ; Canadian Critical Care Trials Group. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 7(10):e47559; 2012.
  55. 55. Niess H, von Einem JC, Thomas MN, Michl M, Angele MK, Huss R, Günther C, Nelson PJ, Bruns CJ, Heinemann V. Treatment of advanced gastrointestinal tumors with genetically modified autologous mesenchymal stromal cells (TREAT-ME1): study protocol of a phase I/II clinical trial. BMC Cancer. 15:237; 2015.

Written By

Raymond M. Wong and Robert B. Cameron

Submitted: 02 June 2016 Reviewed: 24 October 2016 Published: 26 April 2017