Abstract
The therapeutic potential of the immune system in the context of hematologic malignancies has long been appreciated particularly due to the curative impact of allogeneic hematopoietic stem cell transplantation. The role of immune system in shaping the biology and evolution of these tumors is now well recognized. While the contribution of the immune system in anti-tumor effects of certain therapies such as immune-modulatory drugs and monoclonal antibodies active in hematologic malignancies is quite evident, the immune system has also been implicated in anti-tumor effects of other targeted therapies. The horizon of immune-based therapies in hematologic malignancies is rapidly expanding with promising results from immune-modulatory drugs, immune-checkpoint blockade and adoptive cellular therapies, including genetically-modified T cells. Hematologic malignancies present distinct issues (relative to solid tumors) for the application of immune therapies due to differences in cell of origin/developmental niche of tumor cells, and patterns of involvement such as common systemic involvement of secondary lymphoid tissues. This article discusses the rapidly changing landscape of immune modulation in hematologic malignancies and emphasizes areas wherein hematologic malignancies present distinct opportunities for immunologic approaches to prevent or treat cancer.
Distinct aspects of hematologic malignancies as relevant to immune-modulation
In recent years, immunomodulatory approaches have attracted much attention in solid tumors. In this review, we discuss key aspects of the emerging field of immunomodulation, as they apply to hematologic malignancies. Hematologic malignancies are a diverse group of at least 30 different tumor types, each with distinct biology, pathogenesis and clinical behavior. When considering the cross-talk between malignancy and the immune system, it is useful to classify hematologic tumors based on cell of origin, into those that involve multi-potent stem cells (such as acute leukemias), and those (such as lymphoma, myeloma) wherein the target of transformation is a committed lymphoid or myeloid progenitor/progeny. A practical implication of this distinction is that the former are often characterized by severe cytopenias including normal immune cells, and therefore immune-therapeutic strategies need to first restore normal hematopoiesis or adoptively transfer immune effector cells.
Some aspects of the biology of hematologic malignancies deserve distinct considerations (Table 1). Several hematologic malignancies are characterized by systemic involvement of secondary lymphoid tissues. In some settings, the tumor cells may share the developmental niche with normal immune cells leading to immune-paresis. This is commonly exemplified in B cell malignancies such as myeloma and chronic lymphocytic leukemia with a reduction in normal B/plasma cells and consequently hypogammaglobulinemia. In some tumors, cross-talk with normal immune cells may impact tumor growth and survival more directly as it resembles physiologic cell-cell interactions. An example of the latter may be interactions between dendritic cells and tumor cells, or between certain T cell subsets such as T-follicular helper (TFH) cells and tumor cells1,2. Such interactions may also impact immune therapies. For example, TFH cells are characterized by the expression of programmed-death-1 (PD-1), which is also targeted in immune checkpoint blockade.
Table 1.
Some distinct considerations for immune-modulation in hematologic malignancies
Property of hematologic malignancies | Implications |
---|---|
Common involvement of secondary lymphoid tissue | Differences in chemokine requirement for T cell infiltration into tumor tissue |
Shared niche with normal lymphoid cells (lymphoid tumors) | Immune paresis |
Hematopoietic stem cell involvement (e.g. acute leukemia) | Cytopenias |
Role of physiologic interactions with other immune cells due to immune cell of origin | e.g. TFH-B cell interactions may promote tumor growth |
Bone marrow involvement | Marrow as a distinct immune tissue |
Susceptible to both adaptive and innate lymphocytes | Targets for both innate and adaptive immunity |
Precursor states (e.g. MGUS) unresectable | Models to study immune-surveillance |
Less mutational complexity (compared to most solid tumors) | ? more amenable to personalized immunotherapy |
Bone marrow involvement is a common feature of hematologic malignancies. Bone marrow represents a distinct immunologic tissue and therefore properties of marrow-resident immune cells are likely to impact immune therapies against these tumors3. From a genetic perspective, hematologic malignancies carry fewer mutations and exhibit lower degree of intraclonal diversity compared to many solid tumors. These considerations make these tumors attractive targets for therapies targeting selected driver mutations, but the lower mutation load may in principle also translate to fewer neo-antigens and therefore fewer targets for adaptive immune responses. Nevertheless, most hematologic malignancies are highly susceptible to lysis by both innate and adaptive immune cells.
Finally, several hematologic malignancies also represent important models to understand immune-surveillance of human tumors as they are characterized by well-defined precursor states amenable to prospective analysis and the ability to isolate individual tumor/precursor cells and surrounding immune cells. Such serial/prospective studies are typically not feasible in solid tumors because precursor states are resected at initial diagnosis. An excellent example in this regard is monoclonal gammopathy of undetermined significance (MGUS) as a precursor to multiple myeloma (MM).
Immune microenvironment in hematologic malignancies- a dynamic regulator
Several lines of evidence point to an important role for the immune tumor micro-environment (TME) in regulating the growth of hematologic malignancies. For example, prior studies have shown that the immune system has the capacity to recognize the earliest/precursor lesions such as MGUS4. Interestingly, naturally occurring response against MGUS includes antigens commonly expressed on stem cells5. However, even in the setting of clinical myeloma, TME contains T cells capable of killing tumor cells6,7. Therefore mechanisms that suppress preexisting immunity are a critical barrier to immune-mediated destruction. As in solid tumors, these pathways include several mechanisms including myeloid-derived suppressor cells (MDSCs), regulatory T cells (Treg), immunosuppressive cytokines and expression of co-inhibitory checkpoints8. In addition to mechanisms within the TME that inhibit T-cell function, intra-tumoral macrophages and certain subsets of dendritic cells (DCs) can promote tumor growth and even genomic instability9,10.
Properties of the TME also have a major impact on the outcome in other lymphoid tumors including non-hodgkin lymphoma (NHL) and Hodgkin disease (HD). For example, a T-helper1 (Th1) gene expression profile portends improved survival, while a signature of myeloid cells correlates with adverse outcome11. The importance of TME is particularly evident in HD, wherein the tumor cells constitute only a small minority of the tumor bulk and the degree of infiltration of the tumor by T cells or macrophages has a major impact on prognosis12,13. The prognostic impact of TME likely applies across several genetic subtypes.
The potential importance of immune cross-talk in lymphoid tumors is also supported by the findings of recurrent mutations or genomic alterations in major histocompatibility complex (MHC) or key immune genes including those involved in immune checkpoint blockade, which may promote immune ‘escape’14–16. As lymphoid tumors represent tumors involving the immune cells themselves, it is also likely that some of the interactions between immune cells and tumor cells follow physiologic principles. For example, cross-talk between CD28 on tumor cells and CD80/86 on DCs promotes survival of MM cells17. Indeed, signaling via the B cell receptor has emerged as an important regulator of lymphomagenesis and target of newer therapies. Understanding the nature of antigens that drive B cell tumors remains an important area of research and may help elucidate their underlying etiology18.
Several paths to immune-modulation
Allogeneic hematopoietic stem cell transplantation represents perhaps the oldest clinically successful immunotherapy for hematologic malignancies and remains the only curative option for several patients with leukemia. Understanding how to reduce the adverse effects of allo-reactivity (such as graft versus host disease) while preserving anti-tumor graft-versus-leukemia effect remains an area of active research in this field19. Herein we provide an overview of existing/emerging approaches that depend (at least in part) on harnessing the properties of the patient’s own immune system against their own tumors. These approaches can be broadly classified as immune-modulatory drugs/cytokines, vaccines, monoclonal antibodies, immune checkpoint blockade (ICB)/costimulatory antibodies, and adoptive transfer of genetically modified immune cells including chimeric antigen receptor T (CART) cells. It is also apparent that modulation of the immune system may contribute to anti-tumor effects of other non-immunologic therapies, including kinase inhibitors and epigenetic therapies.
Immune-modulating drugs
Thalidomide was initially studied for its immunomodulatory properties, including the capacity to co-stimulate T cells20 and subsequent discovery of its anti-angiogenic effects prompted its evaluation in cancer. Thalidomide and its analogues, notably lenalidomide and pomalidomide have emerged as highly active agents in several hematologic malignancies, and introduction of these therapies has transformed the clinical outlook in MM over the last decade21. Although it is likely that these drugs act by several mechanisms including direct anti-tumor effect and anti-angiogenesis, recent studies have demonstrated the potent capacity of these agents to mediate broad immune activation in vivo, including both innate and adaptive immunity22,23. The underlying mechanisms have been recently ascribed to drug-induced degradation of Ikaros via cereblon-E3 ligase complex, which in turn releases the Ikaros-mediated transcriptional repression on immune activation24–26. In other words, these drugs seem to act in a fashion similar to “checkpoint blockade”, albeit via targeting “intracellular brakes”. Immunostimulatory capacity of these agents includes effects on innate lymphocytes such as natural killer (NK) or NK-T cells22,27. This property likely contributes to the capacity of these agents to enhance antibody-dependent cytotoxicity (ADCC)28 and has led to several clinical trials combining these agents and monoclonal antibodies with promising results, such as in combination with rituximab29. This class of agents also provide proof of principle that immune modulation can be achieved by targeting intracellular molecules implicated in dampening the immune response.
Monoclonal Antibodies (mAbs)
mAbs are multifaceted agents that mediate anti-tumor effects by several mechanisms including direct anti-tumor effects, Fc-mediated immunologic effects such as ADCC, complement-mediated cytotoxicity (CMC), and enhanced antigen-presentation30. Rituximab (anti-CD20 mAb) has transformed the management of B cell malignancies. Preclinical studies have indicated that anti-tumor effects of rituximab require the Fc component31. Newer antibodies targeting CD20 (Ofatumumab and Obinutuzumab) have also shown clinical activity and received FDA approval for management of CLL patients32,33. Ofatumumab is a humanized antibody that binds to a different CD20 epitope than rituximab and mediates greater ADCC32. Obinutuzumab is a glycoengineered antibody that does not engage CMC, but leads to greater ADCC and direct anti-tumor effects34. In addition to CD20, several other targets are currently being explored in the setting of lymphoma. Although early studies with mAbs in the setting of myeloma were disappointing, two antibodies/targets have shown promise. Elotuzumab targets a molecule SLAMF7, which is expressed on myeloma cells, and at a lower level on NK cells35. Based on high response rates in phase II setting, elotuzumab in combination with lenalidomide and dexamethasone is now in phase III testing35. Daratumumab targets CD38 molecule expressed on myeloma cells, and to a lesser extent on T cells36. Daratumumab is one of the first antibodies to show single agent activity in relapsed/refractory MM and received breakthrough designation from the FDA. Two other mAb-based therapies have activity in hematologic malignancies. Brentuximab vedotin is an antibody-drug-conjugate (ADC) that targets CD30 expressed on Hodgkin Reed Sternberg cells and tumor cells in anaplastic large cell lymphoma (ALCL)37. Based on high degree of clinical activity in relapsed HD, Brentuximab received FDA approval in this setting. An important emerging class of agents are bi-specific T cell-engaging (BITE) antibodies that promote physical coupling between tumor cells and T cells. Blinatumomab is a BITE antibody targeting CD3/CD19 that has shown clinical activity in patients with acute lymphocytic leukemia (ALL) and received breakthrough therapy designation by the FDA for relapsed/refractory Philadelphia chromosome-negative ALL38. Both ADC and BITE technologies are very active areas of investigation.
Vaccines
Vaccines have the theoretical advantage of more tumor-specific targeting of tumor cells, thereby reducing the risk of autoimmunity. As the clonal immunoglobulin (Ig) made by each B lymphoid tumor represents the most tumor specific target, several studies have targeted the Ig-associated idiotypic determinants to boost tumor–specific immunity in the setting of lymphoma and myeloma. Early phase studies with this approach led to promising results. However, only one of three phase III trials led to improved progression-free survival39,40. The role of idiotype-based vaccination in the setting of rituximab-based therapies remains to be clarified and is being considered for patients with follicular lymphoma achieving complete remission following induction therapy. Several other tumor-associated antigens are being explored. These include cancer-testis antigens, oncoproteins (such as WT1), and stem cell associated antigens (such as SOX2)5,41,42.
An important set of antigens are those derived from tumor-associated mutations43. Dendritic cells are potent antigen-presenting cells and have been utilized to stimulate T cell immunity as well as innate lymphocytes with promising results in early phase studies23,44. Another emerging approach is antibody-mediated targeting of DCs in situ, which needs to be tested in hematologic malignancies45. Although several of the vaccine-based approaches were effective at inducing immune responses detected in circulation, the clinical efficacy has been modest. Therefore it is likely that such approaches may need to be combined with strategies that target immune-suppressive elements in the tumor bed. As stem cell transplantation (SCT) has been commonly utilized in the setting of hematologic malignancies, the post-SCT setting may be an attractive clinical setting for vaccines or adoptive transfer, as several of the tumor-associated immune tolerance mechanisms may be transiently disabled at that time46.
Immunologic effects of anti-cancer therapies
The possible role of immune system in existing and emerging therapies is now increasingly appreciated. For example, approved therapies such as anthracyclines and bortezomib induce immunogenic death of tumor cells via exposure of heat-shock proteins47,48. In preclinical models, the immune system plays an important role in the anti-tumor effects of alkylating agents, as well as epigenetic therapies commonly utilized against hematologic malignancies49,50. Recent studies have suggested that B cell receptor targeting agents such as Ibrutinib also activate T cells via “off target” effects on ITK51. Finally, oncolytic virotherapies are now targeting hematologic malignancies and in several instances (as exemplified by measles virus targeting myeloma), the immune system may contribute to anti-tumor effects52. While the dominant effect of all of these agents likely involves direct effects on tumor cells, the contribution of an immune component to their anti-tumor effects provides the basis for combinations with immune-therapies.
Adoptive cellular therapies- genetically modified T cells and beyond
An exciting and rapidly emerging area in terms of immunotherapy of hematologic malignancies is the application of adoptive transfer of immune cells, including chimeric antigen-receptor+ T (CART) cells53. Principles of adoptive cell therapy were based on the success of infusion of donor lymphocytes in recipients of allogeneic stem cell transplant and virus-specific T cells in Epstein-Barr virus-driven lymphomas. Preclinical studies provided evidence for the role of homeostatic cytokines during the lymphopenic phase in promoting the growth and survival of transferred cells and led to incorporation of these strategies in clinical protocols54. Several studies are now testing T cells genetically modified to express T cell receptors against tumor antigens or chimeric receptors against cell surface antigens. As expected, a critical issue for these therapies is the choice of the antigen being targeted, as cross-reactivity with antigens expressed on normal cells can be highly problematic55. Second-generation CARTs expressing two co-stimulatory molecules have been effective and those with further modifications are now being tested53. CD19-CARTs have emerged as attractive agents in B cell malignancies as CD19 is often expressed on most tumor cells and the expected toxicity of B cell aplasia is manageable56–58.
Early clinical results with CD19-CARTs were based on therapy of patients with relapsed/refractory CLL56, which were impressive as they targeted disease refractory to multiple therapies. However the results have been even more impressive in patients with B-ALL, with response rates exceeding 80%, including in children59–61. In several of these studies, CART cells have been utilized as a bridge to transplantation. Further studies are needed to establish the durability of responses as a single agent, and whether long-term persistence of transferred cells is needed for durable responses. Clinical studies of CART cells have thus far been restricted to a few institutions, particularly as the management of potentially life-threatening cytokine-release syndrome can be challenging.
CART therapy is now being extended to other hematologic malignancies. For example, CART cells targeting CD38, CD138, BCMA or SLAMF7 are being investigated in MM62. In addition to CART cells, genetically modified T cells expressing TCRs against tumor antigens (such as NY-ESO1) are also being tested in hematologic tumors63. Finally, as recently evaluated in solid tumors, T cells against mutation-derived neoantigens are also ripe for testing in hematologic malignancies43. As discussed earlier, pediatric and hematologic malignancies represent prime candidates for such an approach due to the lack of genomic complexity as typically found in solid tumors.
In addition to T cells, hematologic malignancies are also very sensitive to innate lymphocytes including NK or NKT cells. Evidence for NK-mediated anti-tumor effects has emerged from allogeneic stem cell transplants wherein AML patients were mismatched for killer-cell immunoglobulin-like receptor (KIR) from their donors, which led to enhanced NK cell reactivity due to lack of KIR suppression64,65. Adoptive transfer of NK cells is being attempted in the setting of relapsed leukemia66,67. Lirilumab is an anti-KIR antibody that has been tested in early phase trials in AML and smoldering MM68,69. In addition to NK cells, other innate lymphocytes have also been targeted in hematologic malignancies with promising results. Tumor cells including those in MM commonly express CD1d and are sensitive to lysis by invariant NKT cells70. Tumor progression in MM is associated with NKT dysfunction70. This has prompted studies of adoptive transfer of DCs loaded with iNKT ligand α-galactosylceramide to activate NKT cells as well as adoptive transfer of NKT cells in human MM71.
Immune checkpoint blockade and immune-activating antibodies
As with solid tumors, dampening of T cell function via engagement of co-inhibitory receptors is increasingly appreciated as a mechanism of T cell dysfunction and immune escape in the tumor bed72. Several pathways including PD1/PD-L1, Tim3, Lag-3 and BTLA have been implicated in suppressing T cell immunity in hematologic malignancies73. Expression of PD-L1 on tumor cells as well as associated antigen presenting cells is a common feature of several hematologic malignancies such as MM, leukemia and lymphoma, and is thought to be driven by diverse mechanisms, including cytokines such as interferon-γ in MM or genetic changes such as amplification of 9p24 in HD tumor cells15,73,74. Although prospective studies correlating the expression of inhibitory molecules with clinical outcome are lacking, the preclinical data provide strong rationale for targeting these checkpoints in hematologic malignancies. Initial studies of immune checkpoint blockade in hematologic malignancies with anti-CTLA4 mAb (ipilimumab) evaluated patients with refractory lymphoma and in the setting of relapse for allogeneic transplant75,76.
More compelling evidence for potential benefit from checkpoint blockade has emerged from studies targeting the PD-1 pathway73. The first anti-PD1 antibody tested in hematologic malignancies was pidilizumab. In a phase II trial in patients with relapsed/refractory diffuse large B cell lymphoma, pidilizumab following autograft led to an objective response rate (ORR) of 51% with complete remission (CR) in 34% of patients77. Combination of pidilizumab and rituximab also led to interesting clinical activity in a small phase II trial of patients with follicular lymphoma (ORR 66%, CR 52%)78. More recently, higher affinity anti-PD-1 antibodies such as nivolumab and pembrolizumab, which were developed initially in solid tumors, have been tested in lymphoid malignancies, with evidence of single agent activity in patients with both NHL and HD79–82. The results for single agent activity of PD-1 blockade are most impressive in the setting of relapsed HD, including ORR 87% (CR17%) with nivolumab and ORR 53% (CR20%) with pembrolizumab81,82. Nivolumab was granted breakthrough designation by the FDA for therapy of HD after failure of stem cell transplant and brentuximab. Nivolumab also produced objective responses in 36% of patients with diffuse large B cell lymphoma and 40% of patients with follicular lymphoma79. Several other antibodies targeting PD-1/PD-L1 pathway are entering clinical testing in hematologic malignancies, both as single agents and in combination with other therapies. Results of these ongoing investigations are eagerly awaited. However it now seems likely that combinations based on immune checkpoint blockade may occupy increasing role in the immunotherapy of hematologic malignancies. As with solid tumors, there is an unmet need for biomarkers to identify patients most likely to benefit from these therapies and identify optimal checkpoint targets or combinations. Recent studies demonstrate that immunologic as well as clinical effects of such combinations may be distinct from blockade of individual checkpoints 83,84.
In addition to therapies that block co-inhibitory molecules, agonistic antibodies that enhance immune effector cells are also of interest. It is possible that these antibodies may have distinct toxicity profiles, as evidenced by hepatotoxicity observed in early clinical trials targeting CD13785. Thus far, most of the studies targeting these agents have focused on solid tumors, although preclinical studies also provide a rationale for targeting hematologic malignancies. For example, in lymphoma models, agonistic anti-CD137 antibodies were found to synergize with rituxumab via enhanced ADCC86,87.
Summary and future directions
Therapeutic potential of the immune system against hematologic malignancies has long been appreciated due to the curative impact of allogeneic stem cell transplantation. Hematologic malignancies present several distinct considerations relative to solid tumors for application of immune-modulation. Recent advances in cancer immunology have ushered in a new era with a growing arsenal of several immune-based therapies showing promising clinical activity against hematologic malignancies. Although there are examples of success stories in all categories, much of the recent excitement relates to the promise of ICB and CART therapies.
It is likely that many of these will need to be tested in combination with other existing agents. Integrating these therapies in the clinical management of patients may help realize the potential of the immune system to prevent or treat hematologic malignancies.
Acknowledgments
MVD and KMD are supported in part by funds from the NIH.
Footnotes
Financial Disclosure: MVD has received research funding from Celgene. The authors have no other conflict of interest to disclose.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Tangye SG, Ma CS, Brink R, Deenick EK. The good, the bad and the ugly - TFH cells in human health and disease. Nat Rev Immunol. 2013;13(6):412–426. doi: 10.1038/nri3447. [DOI] [PubMed] [Google Scholar]
- 2.Kukreja A, Hutchinson A, Dhodapkar K, et al. Enhancement of clonogenicity of human multiple myeloma by dendritic cells. J Exp Med. 2006;203(8):1859–1865. doi: 10.1084/jem.20052136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Tokoyoda K, Hauser AE, Nakayama T, Radbruch A. Organization of immunological memory by bone marrow stroma. Nat Rev Immunol. 2010;10(3):193–200. doi: 10.1038/nri2727. [DOI] [PubMed] [Google Scholar]
- 4.Dhodapkar MV, Krasovsky J, Osman K, Geller MD. Vigorous premalignancy specific effector T cell response in the bone marrow of patients with preneoplastic gammopathy. J Exp Med. 2003;198:1753–1757. doi: 10.1084/jem.20031030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Spisek R, Kukreja A, Chen LC, et al. Frequent and specific immunity to the embryonal stem cell-associated antigen SOX2 in patients with monoclonal gammopathy. J Exp Med. 2007;204(4):831–840. doi: 10.1084/jem.20062387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Dhodapkar MV, Krasovsky J, Olson K. T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cells. Proc Natl Acad Sci U S A. 2002;99(20):13009–13013. doi: 10.1073/pnas.202491499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Noonan K, Matsui W, Serafini P, et al. Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors. Cancer Res. 2005;65(5):2026–2034. doi: 10.1158/0008-5472.CAN-04-3337. [DOI] [PubMed] [Google Scholar]
- 8.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chauhan D, Singh AV, Brahmandam M, et al. Functional interaction of plasmacytoid dendritic cells with multiple myeloma cells: a therapeutic target. Cancer Cell. 2009;16(4):309–323. doi: 10.1016/j.ccr.2009.08.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Koduru S, Wong E, Strowig T, et al. Dendritic cell-mediated activation-induced cytidine deaminase (AID)-dependent induction of genomic instability in human myeloma. Blood. 2012;119(10):2302–2309. doi: 10.1182/blood-2011-08-376236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dave SS, Wright G, Tan B, et al. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N Engl J Med. 2004;351(21):2159–2169. doi: 10.1056/NEJMoa041869. [DOI] [PubMed] [Google Scholar]
- 12.Greaves P, Clear A, Owen A, et al. Defining characteristics of classical Hodgkin lymphoma microenvironment T-helper cells. Blood. 2013;122(16):2856–2863. doi: 10.1182/blood-2013-06-508044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Steidl C, Lee T, Shah SP, et al. Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N Engl J Med. 2010;362(10):875–885. doi: 10.1056/NEJMoa0905680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Steidl C, Shah SP, Woolcock BW, et al. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature. 2011;471(7338):377–381. doi: 10.1038/nature09754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Green MR, Monti S, Rodig SJ, et al. Integrative analysis reveals selective 9p24. 1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010;116(17):3268–3277. doi: 10.1182/blood-2010-05-282780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Riemersma SA, Jordanova ES, Schop RF, et al. Extensive genetic alterations of the HLA region, including homozygous deletions of HLA class II genes in B-cell lymphomas arising in immune-privileged sites. Blood. 2000;96(10):3569–3577. [PubMed] [Google Scholar]
- 17.Bahlis NJ, King AM, Kolonias D, et al. CD28-mediated regulation of multiple myeloma cell proliferation and survival. Blood. 2007;109(11):5002–5010. doi: 10.1182/blood-2006-03-012542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Baecklund E, Smedby KE, Sutton LA, Askling J, Rosenquist R. Lymphoma development in patients with autoimmune and inflammatory disorders--what are the driving forces? Semin Cancer Biol. 2014;24:61–70. doi: 10.1016/j.semcancer.2013.12.001. [DOI] [PubMed] [Google Scholar]
- 19.MacDonald KP, Shlomchik WD, Reddy P. Biology of graft-versus-host responses: recent insights. Biol Blood Marrow Transplant. 2013;19(1 Suppl):S10–14. doi: 10.1016/j.bbmt.2012.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Haslett PA, Corral LG, Albert M, Kaplan G. Thalidomide costimulates primary human T lymphocytes, preferentially inducing proliferation, cytokine production, and cytotoxic responses in the CD8+ subset. J Exp Med. 1998;187(11):1885–1892. doi: 10.1084/jem.187.11.1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Palumbo A, Anderson K. Multiple myeloma. N Engl J Med. 2010;364(11):1046–1060. doi: 10.1056/NEJMra1011442. [DOI] [PubMed] [Google Scholar]
- 22.Davies FE, Raje N, Hideshima T, et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood. 2001;98(1):210–216. doi: 10.1182/blood.v98.1.210. [DOI] [PubMed] [Google Scholar]
- 23.Richter J, Neparidze N, Zhang L, et al. Clinical regressions and broad immune activation following combination therapy targeting human NKT cells in myeloma. Blood. 2013;121(3):423–430. doi: 10.1182/blood-2012-06-435503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Licht JD, Shortt J, Johnstone R. From anecdote to targeted therapy: the curious case of thalidomide in multiple myeloma. Cancer Cell. 2014;25(1):9–11. doi: 10.1016/j.ccr.2013.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gandhi AK, Kang J, Havens CG, et al. Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN) Br J Haematol. 2014;164(6):811–821. doi: 10.1111/bjh.12708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kronke J, Udeshi ND, Narla A, et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 2014;343(6168):301–305. doi: 10.1126/science.1244851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chang DH, Liu N, Klimek V, et al. Enhancement of ligand dependent activation of human Natural Killer T cells by Lenalidomide: Therapeutic Implications. Blood. 2006;108(2):618–621. doi: 10.1182/blood-2005-10-4184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tai YT, Li XF, Catley L, et al. Immunomodulatory drug lenalidomide (CC-5013, IMiD3) augments anti-CD40 SGN-40-induced cytotoxicity in human multiple myeloma: clinical implications. Cancer Res. 2005;65(24):11712–11720. doi: 10.1158/0008-5472.CAN-05-1657. [DOI] [PubMed] [Google Scholar]
- 29.Vitolo U, Chiappella A, Franceschetti S, et al. Lenalidomide plus R-CHOP21 in elderly patients with untreated diffuse large B-cell lymphoma: results of the REAL07 open-label, multicentre, phase 2 trial. Lancet Oncol. 2014;15(7):730–737. doi: 10.1016/S1470-2045(14)70191-3. [DOI] [PubMed] [Google Scholar]
- 30.Weiner LM, Dhodapkar MV, Ferrone S. Monoclonal antibodies for cancer immunotherapy. Lancet. 2009;373(9668):1033–1040. doi: 10.1016/S0140-6736(09)60251-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat Med. 2000;6(4):443–446. doi: 10.1038/74704. [DOI] [PubMed] [Google Scholar]
- 32.Wierda WG, Kipps TJ, Mayer J, et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J Clin Oncol. 2010;28(10):1749–1755. doi: 10.1200/JCO.2009.25.3187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Goede V, Fischer K, Busch R, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014;370(12):1101–1110. doi: 10.1056/NEJMoa1313984. [DOI] [PubMed] [Google Scholar]
- 34.Beers SA, Chan CH, French RR, Cragg MS, Glennie MJ. CD20 as a target for therapeutic type I and II monoclonal antibodies. Semin Hematol. 2010;47(2):107–114. doi: 10.1053/j.seminhematol.2010.01.001. [DOI] [PubMed] [Google Scholar]
- 35.Lonial S, Vij R, Harousseau JL, et al. Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. J Clin Oncol. 2012;30(16):1953–1959. doi: 10.1200/JCO.2011.37.2649. [DOI] [PubMed] [Google Scholar]
- 36.de Weers M, Tai YT, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011;186(3):1840–1848. doi: 10.4049/jimmunol.1003032. [DOI] [PubMed] [Google Scholar]
- 37.Younes A, Bartlett NL, Leonard JP, et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363(19):1812–1821. doi: 10.1056/NEJMoa1002965. [DOI] [PubMed] [Google Scholar]
- 38.Topp MS, Gokbuget N, Zugmaier G, et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood. 2012;120(26):5185–5187. doi: 10.1182/blood-2012-07-441030. [DOI] [PubMed] [Google Scholar]
- 39.Schuster SJ, Neelapu SS, Gause BL, et al. Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma. J Clin Oncol. 2011;29(20):2787–2794. doi: 10.1200/JCO.2010.33.3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Freedman A, Neelapu SS, Nichols C, et al. Placebo-controlled phase III trial of patient-specific immunotherapy with mitumprotimut-T and granulocyte-macrophage colony-stimulating factor after rituximab in patients with follicular lymphoma. J Clin Oncol. 2009;27(18):3036–3043. doi: 10.1200/JCO.2008.19.8903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Van Tendeloo VF, Van de Velde A, Van Driessche A, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci U S A. 2010;107(31):13824–13829. doi: 10.1073/pnas.1008051107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Keilholz U, Letsch A, Busse A, et al. A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS. Blood. 2009;113(26):6541–6548. doi: 10.1182/blood-2009-02-202598. [DOI] [PubMed] [Google Scholar]
- 43.Robbins PF, Lu YC, El-Gamil M, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med. 2013;19(6):747–752. doi: 10.1038/nm.3161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 2014;15(7):e257–267. doi: 10.1016/S1470-2045(13)70585-0. [DOI] [PubMed] [Google Scholar]
- 45.Dhodapkar MV, Sznol M, Zhao B, et al. Induction of Antigen-Specific Immunity with a Vaccine Targeting NY-ESO-1 to the Dendritic Cell Receptor DEC-205. Sci Transl Med. 2014;6(232):232ra251. doi: 10.1126/scitranslmed.3008068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lin Y, Zhang L, Cai AX, et al. Effective posttransplant antitumor immunity is associated with TLR-stimulating nucleic acid-immunoglobulin complexes in humans. J Clin Invest. 2011;121(4):1574–1584. doi: 10.1172/JCI44581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Obeid M, Tesniere A, Ghiringhelli F, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13(1):54–61. doi: 10.1038/nm1523. [DOI] [PubMed] [Google Scholar]
- 48.Spisek R, Charalambous A, Mazumder A, et al. Bortezomib enhances dendritic cell (DC) mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications. Blood. 2007;109(Jun 1):4839–4845. doi: 10.1182/blood-2006-10-054221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and immunity. Cell. 2010;140(6):798–804. doi: 10.1016/j.cell.2010.02.015. [DOI] [PubMed] [Google Scholar]
- 50.West AC, Smyth MJ, Johnstone RW. The anticancer effects of HDAC inhibitors require the immune system. Oncoimmunology. 2014;3(1):e27414. doi: 10.4161/onci.27414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dubovsky JA, Beckwith KA, Natarajan G, et al. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood. 2013;122(15):2539–2549. doi: 10.1182/blood-2013-06-507947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Russell SJ, Federspiel MJ, Peng KW, et al. Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clin Proc. 2014;89(7):926–933. doi: 10.1016/j.mayocp.2014.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Maus MV, Grupp SA, Porter DL, June CH. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood. 2014;123(17):2625–2635. doi: 10.1182/blood-2013-11-492231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med. 2005;202(7):907–912. doi: 10.1084/jem.20050732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Linette GP, Stadtmauer EA, Maus MV, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122(6):863–871. doi: 10.1182/blood-2013-03-490565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–733. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368(16):1509–1518. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra138. doi: 10.1126/scitranslmed.3005930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2014 doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra225. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Maus MV, June CH. CARTs on the road for myeloma. Clin Cancer Res. 2014;20(15):3899–3901. doi: 10.1158/1078-0432.CCR-14-0721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sommermeyer D, Conrad H, Kronig H, et al. NY-ESO-1 antigen-reactive T cell receptors exhibit diverse therapeutic capability. Int J Cancer. 2013;132(6):1360–1367. doi: 10.1002/ijc.27792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ruggeri L, Capanni M, Urbani E, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295(5562):2097–2100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
- 65.Venstrom JM, Pittari G, Gooley TA, et al. HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1. N Engl J Med. 2012;367(9):805–816. doi: 10.1056/NEJMoa1200503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bachanova V, Cooley S, Defor TE, et al. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood. 2014;123(25):3855–3863. doi: 10.1182/blood-2013-10-532531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bachanova V, Miller JS. NK cells in therapy of cancer. Crit Rev Oncog. 2014;19(1–2):133–141. doi: 10.1615/critrevoncog.2014011091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Benson DM, Jr, Hofmeister CC, Padmanabhan S, et al. A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood. 2012;120(22):4324–4333. doi: 10.1182/blood-2012-06-438028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Vey N, Bourhis JH, Boissel N, et al. A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission. Blood. 2012;120(22):4317–4323. doi: 10.1182/blood-2012-06-437558. [DOI] [PubMed] [Google Scholar]
- 70.Dhodapkar MV, Geller MD, Chang DH, et al. A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J Exp Med. 2003;197(12):1667–1676. doi: 10.1084/jem.20021650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chang DH, Osman K, Connolly J, et al. Sustained expansion of NKT cells and antigen-specific T cells after injection of {alpha}-galactosyl-ceramide loaded mature dendritic cells in cancer patients. J Exp Med. 2005;201(9):1503–1517. doi: 10.1084/jem.20042592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immune modulation in cancer with antibodies. Annu Rev Med. 2014;65:185–202. doi: 10.1146/annurev-med-092012-112807. [DOI] [PubMed] [Google Scholar]
- 73.Bryan LJ, Gordon LI. Blocking tumor escape in hematologic malignancies: The anti-PD-1 strategy. Blood Rev. 2014 doi: 10.1016/j.blre.2014.09.004. [DOI] [PubMed] [Google Scholar]
- 74.Liu J, Hamrouni A, Wolowiec D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 2007;110(1):296–304. doi: 10.1182/blood-2006-10-051482. [DOI] [PubMed] [Google Scholar]
- 75.Ansell SM, Hurvitz SA, Koenig PA, et al. Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clin Cancer Res. 2009;15(20):6446–6453. doi: 10.1158/1078-0432.CCR-09-1339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bashey A, Medina B, Corringham S, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009;113(7):1581–1588. doi: 10.1182/blood-2008-07-168468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Armand P, Nagler A, Weller EA, et al. Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial. J Clin Oncol. 2013;31(33):4199–4206. doi: 10.1200/JCO.2012.48.3685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Westin JR, Chu F, Zhang M, et al. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. Lancet Oncol. 2014;15(1):69–77. doi: 10.1016/S1470-2045(13)70551-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lesokhin AM, Ansel SM, Armand P, et al. Preliminary results of a phase I study of nivolumab (BMS-936558) in patients with relapsed or refractory lymphoid malignancies. (meeting abstract) Blood. 2014:291. [Google Scholar]
- 80.Armand P, Ansell SM, Lesokhin AM, et al. Nivolumab in patients with relapsed or refractory hodgkin lymphoma- preliminary safety, efficacy and biomarker results of a phase I study. (meeting abstract) Blood. 2014:289. [Google Scholar]
- 81.Moskowitz C, Ribrag V, Michot JM, et al. PD-1 blockade with monoclonal antibody Pembromizumab (MK-3475) in patients wth classical hodgkins lymphoma after brentuximab vedotin failure: preliminary results from a phase Ib study (KEYNOTE-013) Blood. 2014:290. Meeting abstract. [Google Scholar]
- 82.Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 Blockade with Nivolumab in Relapsed or Refractory Hodgkin’s Lymphoma. N Engl J Med. 2014 doi: 10.1056/NEJMoa1411087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369(2):122–133. doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Das R, Verma R, Sznol M, et al. Combination Therapy with Anti-CTLA-4 and Anti-PD-1 Leads to Distinct Immunologic Changes In Vivo. J Immunol. 2014 doi: 10.4049/jimmunol.1401686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Dubrot J, Milheiro F, Alfaro C, et al. Treatment with anti-CD137 mAbs causes intense accumulations of liver T cells without selective antitumor immunotherapeutic effects in this organ. Cancer Immunol Immunother. 2010;59(8):1223–1233. doi: 10.1007/s00262-010-0846-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Houot R, Kohrt H, Levy R. Boosting antibody-dependant cellular cytotoxicity against tumor cells with a CD137 stimulatory antibody. Oncoimmunology. 2012;1(6):957–958. doi: 10.4161/onci.19974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kohrt HE, Houot R, Goldstein MJ, et al. CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies. Blood. 2011;117(8):2423–2432. doi: 10.1182/blood-2010-08-301945. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]