Abstract
While recent progress has been made in the management of multiple myeloma, it remains a highly fatal malignancy especially among patients with relapsed-refractory disease. Immunotherapy with adoptive T cells targeting myeloma-associated antigens are at various stages of development and have brought about a new hope for cure. This is a review on the emerging field of adoptively transferred engineered T cell based approaches, with an in-depth focus on chimeric antigen receptors (CAR) targeting multiple myeloma. The recent results from CAR T cells targeting B cell maturation antigen are encouraging but eventual resistance to the CAR T cell therapies remain problematic. With newer approaches in therapies for multiple myeloma, the role of transplantation is evolved to form a platform for T cell therapies.
Keywords: Multiple myeloma, chimeric antigen receptors, BCMA, Hematopoietic cell transplantation, CAR, immunotherapy, TCR
Introduction
Multiple myeloma (MM) is the second most common hematologic malignancy in the United States and is associated with high rates of fatality [1]. Recent years have seen an emergence of newer drugs and regimens to treat MM with response rates approaching 100% [1-5]. These regimens provide a safer and more durable option for disease control in most patients with MM. In a small percentage of patients, conventional therapies might be curative by themselves [6,7]. However, in the majority these therapies are not curative and most patients with MM die from their disease. Thus, the quest for potentially curative but safer options continues. This is especially relevant for patients with relapsed or refractory MM.
Immunotherapy has revolutionized the landscape of cancer therapies in recent years [8]. This revolution has been powered by the parallel development of newer ways to reinvigorate endogenous tumor-reactive cells with immune check point blockades [9] and engineering of T cells ex vivo for adoptive transfer [10]. This has positively influenced the outlook for therapeutics of MM and heralded exploratory translational and clinical research focused to achieving improved responses in relapsed-refractory MM.
Allogeneic hematopoietic cell transplantation in MM
Allogeneic hematopoietic cell transplantation (alloHCT) is the first proven curative treatment for MM [11]. It is a viable option for select patients with relapsed-refractory MM in whom potential benefits of the procedure outweigh its risks and alternate options. The success of alloHCT in MM has been tempered by historical high rates of non-relapse mortality (NRM) which can reach 40% and adversely impacts overall survival [12]. NRM is primarily caused by infections due to prolonged immune suppression and graft versus host disease (GVHD), a broad immune attack of the donor T cells against normal host tissue.
Improvements in the process of alloHCT have, however, led to significant improvements in NRM. For example, using myeloablative conditioning and CD34+ selected allografts in 44 patients treated at MSKCC from 2007-2013 one-year NRM rates of 18% and grade II-IV acute and chronic GVHD rates of 2% and 0% respectively, were obtained [13]. In spite of improvements in NRM rates of MM, relapse remains a prominent problem in patients with relapsed-refractory disease. In our institutional experience, despite the use of high intensity conditioning to eradicate disease, in this heavily pre-treated, high risk population, progression free survival (PFS) in alloHCT recipients had a median duration 13.5 months and a PFS rate of 31% at 2 years, although a “tail of the curve” was seen with a significant minority of long term progression free survivors out to 6 years post-alloHCT.
This speaks to the curative potential of alloHCT, which is based on a combination of high intensity conditioning to eradicate disease, and in contrast to autologous HCT (autoHCT), reinfusion of an allogeneic hematopoietic cell source to induce anti-MM immunity. A graft-versus-myeloma effect is achieved where the T cells derived from the donor graft recognizes the recipient-derived malignant cells as foreign, and therefore targets, and eliminates them [14]. Thus alloHCT, despite it toxicities, remains the most established form of T cell based immunotherapy in MM, and serves as evidence for the curative potential mediated by the T cell recognition and lysis of MM cells. Multiple groups have been investigating enhancing anti-MM T cell responses, while simultaneously improving the safety of such approaches, through adoptive gene transfer of either a synthetic T cell receptor (TCR) or a chimeric antigen receptor (CAR) and re-infusion of autologous gene modified T cells.
Adoptive transfer of T cells for myeloma
Anti-myeloma immunity participates in controlling MM, and a compromised immune system is associated with more aggressive disease [15-17]. T cells reactive to myeloma-associated antigens are detectable and correlate with disease burden [18]. Prognostically significant T cell clones are also seen following autoHCT [19] and alloHCT [20]. These observations encourage the development of adoptively transferred T cell-based strategies to treat MM.
Endogenous T cell activation and reinfusion
It has been suggested that reduced intensity or non-myeloablative conditioning in combination with delayed adoptive transfer of donor T cells, in the form of donor lymphocyte infusion (DLI), can lead to retained graft-versus-myeloma effects while mitigating GVHD and overall NRM [21]. In an early attempt, four patients with advanced MM were treated with DLI leading to responses in three of the patients and a conversion of hematopoietic chimerism to donor-derived [22]. In spite of the response and full donor chimerism, the remission was not durable and relapse occurred. A subsequent dose escalation trial of DLI included 19 MM patients after alemtuzumab-related conditioning [23]. Of these patients, 12 had only modest response, which were associated with GVHD. With advent of newer agents, combination therapies with DLI have been attempted. Combination of bortezomib and dexamethasone prior to DLI showed only modest responses and co-occurrence of GVHD [24]. In another trial, lenalidomide was used as maintenance after DLI and showed mildly better responses and PFS compared to lenalidomide maintenance alone [25].
Given the difficulty of separating graft-versus-myeloma and GVHD with unmodified allogeneic T cells, adoptive transfer of ex vivo processed autologous T cells have been studied. Success of adoptively transferred T cell therapy is dependent on generation of myeloma-specific T cell clones that can be expanded ex vivo, proliferate and kill target cells in vivo in myeloma niches, and attain a long-lasting memory state for continued immune surveillance [26]. One strategy to obtain a large number of tumor specific T cells is to expand tumor-infiltrating lymphocytes (TILs) and adoptively transfer the expanded cells back to the patient. Indeed TILs have been used with measurable success in solid tumors like melanoma [27]. In MM, when compared to peripheral blood lymphocytes, bone marrow-infiltrating lymphocytes (MILs) have an increased likelihood of a precursor T-cell population with an enhanced tumor specificity [28]. Activated MILs (aMILs) were adoptively transferred after in vitro stimulation and expansion for 7 days in 22 patients with relapsed-refractory MM [29]. Patients were treated with high dose melphalan, autologous hematopoietic cell rescue, and infusion of aMILs. Among the treated patients, the overall clinical response was 54% with 27% complete response (CR, no paraprotein in urine or serum and <5% BM plasma cells) and 27% partial response (PR, >50% reduction in paraprotein). Remarkably, demonstrable myeloma-specific T cells responses were not uniformly observed in all the treated patients, but the presence of a myeloma-specific immune response was associated with clinical response.
TCR gene transfer for MM
To obtain a more uniform myeloma-specific response, myeloma-associated antigens have been targeted to generate redirected T cells for adoptive transfer. Several myeloma associated antigens have been identified. MAGE A3, which is a cancer-testis antigen (CTA), is expressed in myeloma cells. T cells have been genetically engineered to express an affinity enhanced synthetic TCR (sTCR) directed to a MAGE A3 antigen-derived peptide/human leukocyte antigen (HLA)–A*01 complex [30]. However its use has been restricted due to cross-reactivity against a cardiac myocyte associated antigen peptide titin, leading to fatal cardiovascular toxicity in the first two patients treated [31]. NY-ESO-1 is another CTA expressed in multiple cancers [32]. Advanced myelomas express NY-ESO-1 which is highly immunogenic [33]. NY-ESOc259 is an affinity-enhanced sTCR that recognizes the NY-ESO-1–derived peptide in the context of HLA-A*0201 [34]. 20 patients were treated on a trial with high dose melphalan, autologous hematopoietic cell rescue, and subsequent administration of adoptively transferred NY-ESOc259 T cells. Of note, for most of these patients it was their first autologous transplant. 14 had CR or near-CR, 2 patients had a very good partial response (VGPR; ≥90% reduction in paraprotein levels), and 2 had PR. The immune correlates of this adoptive cell therapy trial were promising. NY-ESOc259 T cells home to disease marrow, proliferated, and long-term persistence was seen that correlated with clinical activity. However the efficacy induced by the adoptive T cell therapy by itself in both these cases is confounded by the co-administration of high dose melphalan and autoHCT.
CAR T cell therapy for MM
Fundamental to specificity of naturally occurring and synthetic T cells is the interaction of a cognizant TCR complex with its corresponding peptide presented in HLA complex in the target cells (Figure 1). The requirement of TCR and HLA interaction for T cell effector function, which is unique to each individual, and can be down-regulated in neoplasms, can be overcome by the use of chimeric antigen receptors (CARs) [35]. Furthermore, CAR modified T cells do not rely on endogenous activation and co-stimulation but receive supra-physiologic stimulatory signals through the CAR. Chimeric antigen receptors are synthetic fusion proteins consisting of the variable portion of antibody, known as a single chain variable fragment (scFv) that can target an antigen displayed on the surface of a tumor cell [36]. The basic construct of the CAR gene encodes the scFv in frame with a transmembrane domain, and T cell activation and co-stimulatory signaling domains from the TCR CD3 zeta chain, and most commonly CD28 or 4-1BB. The synthetic gene is introduced in the T cell using retroviral or lentiviral vectors that ensure genetic integration and constitutive expression.
Figure 1. Activation of TCR and CAR.
The TCR complex of a cytotoxic T cell binds to a peptide presented in the context of a HLA class- I molecule. This immunological synapse triggers a signaling cascade and in the presence of appropriate costimulatory signals cause activation of the cytotoxic T cells. A CAR is a synthetic fusion protein containing a single chain variable fragment (scFv) that can directly bind to target cell surface antigen. This engineered CAR synapse activates the immunoreceptor tyrosine-based activation motif (CD3e-ITAM) and, if present, costimulatory molecules causing activation of the CAR T cell.
CAR T cell therapies targeting CD19 have been the most widely used and successful among all the chimeric antigen receptors to date [37]. CD19 is a surface antigen expressed on the membranes of precursor and mature B cells. It is also expressed on various B cell malignancies including B-acute lymphoblastic lymphoma, chronic lymphocytic leukemia and non-Hodgkin lymphoma [38]. Autologous CD19 CAR T cells have accordingly been used in CD19-expressing B cell malignancies with dramatic success [39-43].
CD19 CAR T cells for MM
MM, a neoplastic disorder of plasma cells, does not routinely express detectable CD19. There are however, several scenarios that can be imagined where patients with MM might benefit from CD19 targeted CAR T cell therapy. First, it may be possible that MM cells express low levels of CD19, below our limit of detection by routine flow cytometric and immunohistochemical approaches, which may be sufficient for CAR-mediated killing. Second, there may be MM progenitor cells, which are B cell in origin and presumably express CD19. Third, there may be regulatory CD19+ B cells that play a supportive/immunosuppressive role in the MM microenvironment, and eliminating this population may allow for an endogenous MM infiltrating lymphocyte anti-tumor effect.
The use of CD19 CAR T cells have been studied as a salvage therapy in MM patients with relapse-refractory myeloma who had previously received autoHCT [44,45]. In this study the patients were treated with high dose melphalan and a second autoHCT, followed two weeks later by CTL019, a lentivirally transduced CD19 CAR T cell product. The maximum target dose of CTL019 was 5×107. The results of the first patient in this study were encouraging and published [44]. The day 100 bone marrow biopsy of the patient showed 1 to 2% overall cellularity with no plasma cells, and stringent complete response was seen. No evidence of cytokine release syndrome was observed. Twelve months after transplantation, the patient had no laboratory evidence, and no clinical signs or symptoms of MM. Surprisingly, at this time point, CTL019 was not detectable in blood and marrow and non-neoplastic CD19+ B cells had reconstituted suggesting that sustained CD19 CAR activity was likely lost or substantially reduced. Since the publication of the initial report, data from a total of 10 patients (inclusive of the patient above) were presented at the 2016 annual meeting of annual society of hematology [45]. Overall, 2 out of the 10 patients including the one presented in detail above, had “remission inversion” or a longer PFS after this second administration of high-dose melphalan. None had an ongoing clinical response. While, a favorable PFS correlated with peak frequency of CTL019 in the bone marrow, there was no association between PFS and peak frequency of CTL019 or duration of CTL019 persistence in peripheral blood.
κ.CAR T cells
One drawback of targeting antigens expressed on all B cells is the development of hypogammaglobinemia leading to profound immunosuppression. Since myeloma cells are monoclonally restricted for κ or λ immunoglobulin subtype, one strategy can be to target the monoclonal subtype of malignant B cells sparing the normal B cells. As a proof of principle, κ.CAR T cells were generated targeting κ light chain (referred to as κ.CAR T cells) [46]. Seven MM patients received conditioning with 12.5 mg/kg cyclophosphamide followed by κ.CAR T cell infusion (9.2×107 to 1.8× 108 κ.CART cells/m2). The responses were modest and of the 7 patients, 4 had responses in the form of stable disease. The responses were substantially better with NHL and CLL. As with CD19 CAR T cells, one obvious drawback of using this approach is that, like healthy plasma cells, myeloma cells secrete and do not retain expression of immunoglobulin on their surface and the anti-myeloma effects would be dependent on targeting of MM precursors.
CD138 CAR T cells
CD138 (syndecan-1) is a surface molecule highly expressed on human plasma cells including myeloma cells [47]. A CD138 targeted CAR including a 4-1BB costimulatory domain was tested in five relapse-refractory MM patients in standard phase one dose-escalation trial with 3–6 planned CD3+ T-cell doses, averaging 7.56 × 106 cells/kg [48]. The best responses were SD in 3 of the patients at 3 months. No intolerable toxicities were observed in this process. This is surprising since CD138 is broadly expressed in human tissues including epithelial cells, vascular smooth muscle cells, and the endothelium [49]. It can be postulated in light of the modest response against myeloma seen in the above trial and the less-than-expected toxicities with CD138 CAR T cells was due to a less potent CAR activity and it is possible that a more potent CAR activity would result in vigorous toxicities.
BCMA CAR T cells
The ideal target antigen in MM for CAR T cell therapy would be a molecule that is highly expressed on the surface of all neoplastic plasma cells. B-cell maturation antigen (BCMA) is an excellent candidate due to its preferential expression in plasma cells [50].
BCMA is a TNF receptor superfamily 17 (TNFRSF17) that plays a central role in regulating B-cell maturation and differentiation into PC [51]. It is a type III transmembrane protein lacking a signal-peptide but containing cysteine-rich extracellular domains, and promotes B-cell survival and differentiation [52]. It is not present on naïve B cells or hematopoietic stem cells, but is upregulated during B-cell differentiation into plasmablasts [50]. It was initially believed that BCMA is not expressed in non-hemopoietic tissue. However recent data suggest that BCMA is abnormally expressed in non-small cell lung cancer cell (NSCLC) lines and may play a role in NSCLC tumors through the ERK1/2 signaling pathway [53]. Fortunately, there is no evidence of the expression of this molecule in normal essential non-hematopoietic tissue.
A first-in-human trial conducted at the NCI used autologous T cells transduced with γ-retroviral vector encoding a BCMA targeted CAR (NCI mBCMA CAR) [54] (Figure 2). The BCMA scFv of the NCI mBCMA CAR is derived from a murine hybridoma and includes a human CD28 co-stimulatory domain. Only patients with BCMA >50% by IHC or flow were treated, and 30% of the patients were excluded based on low expression of BCMA, ensuring more uniform BCMA expression. All patients received cyclophosphamide and fludarabine conditioning chemotherapy prior to CAR T cell infusion. The initial report on the first twelve patients treated in dose escalation cohorts with doses ranging from 0.3×106 to 9×106 CAR T cells/kg showed that two of the three patients treated at the highest dose level (9×106 CAR T cells/kg) showed dramatic responses, including a stringent CR. A dose of 9×106 cells/kg was also associated with grade 3-4 toxicities, most significantly, cytokine release syndrome in both the responding patients.
Figure 2. The components of BCMA-CAR T cells.
A schema depicting the components of the five BCMA targeted CARs currently being tested in clinical trials. NCI (National Cancer Institute); Bluebird (Bluebird Bio Multi-institutional trial of b2121): U Penn (University of Pennsylvania); MSKCC (Memorial Sloan Kettering Cancer Center)
The trial utilizing NCI mBCMA-CAR T cells was the first demonstration of dramatic responses induced by non-CD19 targeted CAR T cell therapy. Subsequently three additional BCMA targeted CAR T cell trials for MM have opened in the US (Figure 2, Table 1). Bluebird mBCMA-CAR, known as bb2121, which grew out of the NCI program is being tested in a multi-institutional trial. Initial data from this trial was presented at 2016 EORTC-NCI-AACR Molecular Targets and Cancer Therapeutics Symposium (J.G. Berdeja 2016, unpublished). The construct is a lentiviral vector encoding a mouse-derived scFv and includes a 4-1BB costimulatory domain. As above, this trial too includes patients with BCMA expression > 50% and all patients receive conditioning chemotherapy with fludarabine and cyclophosphamide. Initial reports of this dose escalation trial reveals that at the dose of 150×106 total T cells (second of three dose levels reported) 3 out of 3 patients had dramatic responses (1 VGPR, 2 sCR) lasting >20 weeks. Interestingly at the third dose level (450×106 total T cells), 3 of the 3 patients treated had only partial responses. Notably, no grade 3/4 toxicity was observed in any patient treated at the first 3 dose levels. The trial continues to enroll at higher dose levels.
Table 1.
Summary of the trial design of clinical trials with BCMA-CAR T cells.
Institution/CAR | NCI mBCMA-CAR | Bluebird mBCMA-CAR (bb2121) | UPenn huBCMA-CAR | MSK huBCMA-CAR | Nanjing Legend LCAR-B38M |
---|---|---|---|---|---|
Clinicaltrials.gov Identifier | NCT02215967 | NCT02658929 | NCT02546167 | NCT03070327 | NCT03090659 |
BCMA Ag screening | >50% | >50% | None | >1% | Clear expression |
Conditioning (Day 0 denotes day of CAR T cell infusion) | Cyclophosphamide and fludarabine on days -5, -4, and -3 | Cyclophosphamide and fludarabine on days -5, -4, and -3 |
Cohort 1: none Cohort 2, 3: Cyclophosphamide on day-3 |
Cohort 1: Cyclophosphamide on day –2 Cohort 2 onwards: Cyclophosphamide and fludarabine on days -4, -3, and -2 |
Cyclophosphamide |
Planned enrollment | 38 | 50 | 27 | 24 | 100 |
University of Pennsylvania has also reported on a dose escalation trial with their BCMA targeted CAR T cell product. The UPenn huBCMA-CAR is a lentiviral construct encoding scFv against BCMA identified from a human library and includes the 4-1BB costimulation domain Initial data from this trial was presented at the 2016 annual meeting of annual society of hematology [55]. Unlike the previous trials discussed, the patients included in this trial are not screened for BCMA expression, and the first cohort treated did not include any conditioning chemotherapy. From this cohort, nine patients have been reported on at a goal 500×106 total CAR T cells. This cohort is notable for a single patient with a sCR ongoing for greater then 12 months. In addition to the sCR, 2 other patients achieved VGPRs. Four of the nine patients reported required anti-IL6 receptor therapy with Tocilizumab for CRS.CAR T cell persistence has been measured up to 24 weeks. The trial is now enrolling patients to be treated with cyclophosphamide conditioning regimen.
It is worthwhile to highlight that the NCI and Blubird bio trials use a murine hybridoma-derived scFv, while the UPenn and the newly opened MSKCC studies utilized a human library screening approach to identify scFv's. Experience from mouse hybridoma-based CD19-CAR study from Fred Hutchinson Cancer center suggests potential advantages with a human library screening approach. It is observed that expansion after repeat administration of CAR T cells is limited in at least some patients by the development of host anti-murine scFv immune responses [56]. Repeated host anti-CAR immune responses may be less against human derived CARs. This may also mitigate the necessity of a strong conditioning regimen as seen in the UPenn huBCMA-CAR trial, where examples of impressive efficacy were noted without the use of prior conditioning chemotherapy. Additionally, a library provides investigators with an expansive selection of scFvs to select a superior lead candidate.
At MSKCC, we have recently opened the fourth phase I trial using human-library identified BCMA scFv (ClinicalTrials.gov Identifier: NCT03070327). The MKSCC huBCMA-CAR is a retroviral construct and includes a 4-1BB costimulatory domain. Patients with MM expressing any levels of BCMA positivity (>1%) are eligible. The conditioning regimen is cyclophosphamide for the first cohort, with patients treated in subsequent cohorts additionally receiving fludarabine.
Besides these trials in the United States, BCMA CAR trails are also open internationally. Investigators from Xi¹an Jiaotong University reported exciting results from a phase I trial of LCAR-B38M anti-BCMA CAR T cells in 34 patients with relapsed/refractory multiple myeloma and two patients with plasma cell leukemia [57]. 100% of the patient reported an objective response with 15 complete responses. The exact nature of the scFv, manufactured by Nanjing Legend is not clear. Of note, the patients in the above trial received CAR T cells after a median of 3 lines of therapy, which is substantially earlier than the other trials described above.
In all the reported trials, resistance to BCMA CAR T cells among a substantial group of patients remains an area of active investigation. It is yet to be understood how BCMA expression (low vs high) impacts the efficacy of BCMA targeted CAR T cell therapy. Different thresholds for eligibility may have an impact on response rates and durability between trials. Once there is mature data, it will be important to consider this factor when choosing therapies for an individual patient. The mechanisms of resistance are still not well understood but the emergence of BCMA-negative myeloma cells and possible requirements of a higher threshold of surface BCMA expression for optimal killing are important clues to unraveling the mystery. In-depth immunological studies of the kinetics of T cells in the myeloma milieu and studies on the expression of BCMA on cell surface in the future trials will be of great importance in understanding the mechanism of resistance to BCMA targeted CAR T cells.
Non-BCMA targeted CAR T cells for MM
In addition to the above targets for CAR based therapies, there are additional candidate targets for CARs being explored in preclinical and very early clinical stages. CD38 (ADP-Ribosyl Cyclase) is type −II glycoprotein and a cell surface antigen structurally related to proteins such as transferrin receptor and the HLA-molecule [58]. It is consistently expressed on malignant plasma cells, making it an attractive target for CAR T cell therapy. It is also expressed on other B cell subtypes and other BM-derived progenitors including myeloid and lymphoid precursors, and subtypes of T cells [59]. Daratumumab is a CD38-targeting monoclonal antibody, approved for treating patient with relapsed-refractory MM [60]. The promising activity of daratumumab in MM has led to the evaluation of CD38-targeting CARs. In preclinical testing, retrovirus targeting CD38 has shown efficacy against patient-derived myeloma lines [61] and in xenograft models [62]. In spite of the promise, CD38 is a less than an ideal CAR target given that its expression in malignant plasma cells is known to decrease and is lost in the most aggressive forms, setting the stage for likelihood of resistance to CD38 CAR [63]. It is also expressed on activated T cells, which might lead to fratricide among CD38 CAR T cells [64]. Moreover, CD38 is also expressed on normal non-hematopoietic tissues including prostate epithelial cells, and while the antibody is safe for use in patients, the more potent potential of CAR T cells does not guarantee safety when targeting this widely expressed protein [65].
Other CAR approaches have included specific targeting of isoforms of widely expressed antigens. The isoform variant 6 (CD44v6) is expressed in a variety of malignant cells including myeloma cells and CAR T cells targeting CD44v6 mediated specific malignant cell killing in xenogenic models while sparing normal CD44+ hematopoietic cells [66]. In another interesting approach, CAR T cells targeting CD70, which is expressed on various B cell malignancies including MM, killed CD70-positive tumor cell lines and primary tumors in vitro and in a murine SCID xenograft model [67]. CD70 is a natural ligand of CD27 and the CAR designed consisted of full-length CD27 as the antigen-recognition domain fused to the intracellular domain of the CD3-ζ chain. The CARs being tested in MM have not been limited to T cells alone. Natural Killer (NK) cells retrovirally transduced with CARs targeting SLAM7 (CS1), a glycoprotein expressed on normal and malignant plasma cells [68], have shown capacity to lyse in vitro and xenogenic models of MM [69].
Future directions
AlloHCT as a platform for T cell therapy in myeloma
In light of the rapidly emerging cell therapy options in MM, the role of alloHCT is in the process of refinement. Most clinical trials with CAR T cells exclude patients who have undergone a prior alloHCT, thus potentially limiting treatment options for patients with high-risk MM who choose to undergo alloHCT early in their disease course and then relapse. Thus, not having the option to receive CAR T cells on clinical trials may influence the patients and their physicians to pursue experimental CAR T cell therapy prior to alloHCT. However, to date, the data is not clear if CAR T cells can replace alloHCT as a curative therapy. Further, the restricted availability of CAR T cell trails and the increasing possibilities of suitable donors of allograft, alloHCT remains relevant in therapies of MM, especially for high risk patients.
Fundamental to the relevance of alloHCT is its potential to cure difficult to treat MM delivered by a combination of high dose chemotherapy, tumor-free hematopoietic cell product, and graft versus myeloma effect. Preclinical studies with alloHCT have been focused on the development of engineered donor cell products that can enhance the antitumor effects across HLA barriers with minimal transplant-related mortality [70]. Studies in mouse models of alloHCT suggest that by optimizing the timing of adoptive transfer and specific targeting of effectors functions of T cells, enhanced antitumor effects of donor T cells can be achieved without enhancing GVHD [71-73]. Thus alloHCT can be developed as a platform for therapy of relapse refractory myeloma for donor-derived CAR and non-CAR T based therapies. Preclinical [74] and clinical data [75,76] with donor-derived CD19 CAR T cells suggest that in an allogeneic setting, concurrent stimulation of CAR and alloreactive TCR can eliminate GVHD-causing donor T cells population while retaining non-alloreactive CAR expressing T cells that can mediate anti-tumor effects.
Recent understanding of T-cell development have fed forward into the prospects of using T-cell progenitor cells to enhance post-transplant immune reconstitution [77]. Since donor T cell progenitors undergo further education in the host thymus, host-directed tolerance develops minimizing the risk of GVHD. T cell progenitors can be further engineered to expressed CAR as an off-the-shelf T cell based therapy agent [78]. In humans, umbilical cord-derived T cell progenitors expressing CARs have shown promise in preclinical studies [79,80] and can be expected to be valuable in developing anti-myeloma cell therapeutics.
Overcoming immune resistance
Although MM is amenable to immunotherapy in the form of alloHCT and myeloma-directed adoptive T cell transfer, development of resistance and the lack of optimal responses remain problematic. In addition to newer target identification for CARs and sTCRs, efforts are also focused on identifying mechanisms of myeloma-intrinsic resistance to T cell mediated killing and engineering T cells and overcoming the resistance.
Combining CAR T cells and immune checkpoint blockades
T-cell exhaustion/senescence is a distinguishing feature of myeloma relapse [81]. As with endogenous T cells, PD1/PDL-1 axis can attenuate myeloma specific T cell activity. PD-L1 is expressed by myeloma cells [82,83] and the myeloma bone marrow induces expression of PD-L1 [84]. As a single agent, PD1 inhibition in relapsed-refractory MM has shown suboptimal responses in two small studies (4% observable response in 27 RRMM patients treated with nivolumab, [85] stable disease in 1RR MM patient treated with pidilizumab [86]). However, the observation of stable disease in 63% [85] in the heavily pretreated relapsed-refractory MM population suggests potential effectiveness in combinatorial settings. Clinical trials are underway to explore the effectiveness of PD-1 and PD-L1 inhibition in combination with proteasome inhibitors and immunomodulatory agents (IMiDs) in pretreated [87] and therapy-naïve MM patients [88]. Reports of early results show significant efficacy in the relapsed-refractory population when Pembrolizumab is combined with an IMiD (50% ORR with lenalidomide plus dexamethasone (KEYNOTE-023), 60% ORR with pomalidomide and dexamethasone (KEYNOTE-183). However due to unforeseen toxicities including deaths, the above trials have placed on hold, which have tempered the enthusiasm for the use of immune checkpoint blockades in MM.
The combination of immune checkpoint blockade with adoptive cellular therapy is nevertheless intriguing. In mouse models of sarcoma and breast cancer, in which the combination of PD-1 blockade with murine CAR T cells showed a significantly enhanced antitumor effects compared with either intervention alone [89]. CRISPR/CAS9 mediated disruption of the immune checkpoints rending CAR T cells resistant to PD1 inhibition has also shown promise in preclinical models [90]. We predict that combination of adoptively transferred T cells and immune checkpoint blockade will prove to be synergistic in human studies.
Armored CAR
MM is known to have a complex immunosuppressive microenvironment consisting of recruited or co-opted immunosuppressive cell types and an immunosuppressive cytokine milieu which impairs activity of proimmune TH1 cells and restricts antitumor T cell immunity [91,92]. While the antigen-specific ability of CAR T cells are being exploited with discoveries of myeloma-specific targets, optimizing the signaling and cytolysis of CAR T cells are yet to be fully explored. Armored CAR T cells represent the next generation of optimization that focuses on reinforcing modified CAR T cells against the influences of an immunosuppressive tumor microenvironment [93,94]. Armored CAR strategies have spanned engineering endogenous cytokine production and enhancing costimulatory effects.
Effects of immune inhibitory TH2 cytokines can be overcome by modifying CAR T cells to secrete IL12 which enhances expression of IFN-γ and IL12 and leads to more effective clearance of tumors in murine models [80,95]. Enhanced effects of CAR T cells have also been shown with transgenic co-expression of IL15 [96] and IL21 [97]. Further development of IL15 expressing CARs were hampered out of concern for leukemogenic potential of IL15 [98]. Transgenic co-expression of IL12 and IL21 remain exciting and unexplored opportunities to enhance efficacy and persistence of myeloma specific CARs.
Thus far, the CAR T cells used in clinical trials and most preclinical studies utilize costimulatory-signaling domains such as CD28 or 4-1BB. The choice of costimulatory domain of the CARs may impact cellular activation, persistence [99,100], and development of immunologic exhaustion [101]. The immunosuppressive myeloma microenvironment can also be overcome using constitutive expression of CD40L, which facilitates cytolysis and proliferation of CAR T cells, thus augmenting tumor immunogenicity [102]. Additionally, improved antigen presentation mediated by constitutive expression of CD40L can recruit endogenous antitumor T cells and cause sustained responses.
Conclusion
Taken together, MM is a disease conducive to adoptive T cell-based therapies. Chimeric antigen receptors are currently being tested in clinical trials and initial results have been encouraging. Newer targets for T cell therapy are being investigated. However, emergence of resistance to cellular therapies due to intrinsic immunosuppressive networks remains a major barrier to achieving cure. Further research into the mechanisms of resistance is crucial to develop strategies to maximize durability of responses while minimizing toxicities associated with cell therapies. Developing appropriate platforms for myeloma targeted cell therapy and designing CAR T cell that are resistant to immunosuppressive microenvironment are exciting avenues for the future on myeloma targeted treatment.
Acknowledgments
AG is supported by NIH/NCATS Grant #UL1TR00457 administered by the Clinical and Translational Science Center at Weill Cornell Medical Center and MSKCC. ELS is supported by LRF, SITC, and an MSK Technology Development Grant. All investigators acknowledge MSK Cancer Center Support Core Grant (P30 CA008748).
References
- 1.Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood. 2008;111:2516–2520. doi: 10.1182/blood-2007-10-116129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mailankody S, Korde N, Lesokhin AM, et al. Minimal residual disease in multiple myeloma: bringing the bench to the bedside. Nat Rev Clin Oncol. 2015;12:286–295. doi: 10.1038/nrclinonc.2014.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kazandjian D, Landgren O. A look backward and forward in the regulatory and treatment history of multiple myeloma: Approval of novel-novel agents, new drug development, and longer patient survival. Semin Oncol. 2016;43:682–689. doi: 10.1053/j.seminoncol.2016.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Korde N, Roschewski M, Zingone A, et al. Treatment With Carfilzomib-Lenalidomide-Dexamethasone With Lenalidomide Extension in Patients With Smoldering or Newly Diagnosed Multiple Myeloma. JAMA Oncol. 2015;1:746–754. doi: 10.1001/jamaoncol.2015.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jakubowiak AJ, Dytfeld D, Griffith KA, et al. A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma. Blood. 2012;120:1801–1809. doi: 10.1182/blood-2012-04-422683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Alexanian R, Delasalle K, Wang M, Thomas S, Weber D. Curability of multiple myeloma. Bone Marrow Res. 2012;2012:916479. doi: 10.1155/2012/916479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Barlogie B, Mitchell A, van Rhee F, Epstein J, Morgan GJ, Crowley J. Curing myeloma at last: defining criteria and providing the evidence. Blood. 2014;124:3043–3051. doi: 10.1182/blood-2014-07-552059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016;13:273–290. doi: 10.1038/nrclinonc.2016.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Sci Transl Med. 2015;7:280s. doi: 10.1126/scitranslmed.3010274. r281. [DOI] [PubMed] [Google Scholar]
- 10.Themeli M, Riviere I, Sadelain M. New cell sources for T cell engineering and adoptive immunotherapy. Cell Stem Cell. 2015;16:357–366. doi: 10.1016/j.stem.2015.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Festuccia M, Martino M, Ferrando F, et al. Allogeneic stem cell transplantation in multiple myeloma: immunotherapy and new drugs. Expert Opin Biol Ther. 2015;15:857–872. doi: 10.1517/14712598.2015.1036735. [DOI] [PubMed] [Google Scholar]
- 12.Gahrton G, Tura S, Ljungman P, et al. Allogeneic bone marrow transplantation in multiple myeloma European Group for Bone Marrow Transplantation. N Engl J Med. 1991;325:1267–1273. doi: 10.1056/NEJM199110313251802. [DOI] [PubMed] [Google Scholar]
- 13.Smith E, Devlin SM, Kosuri S, et al. CD34-Selected Allogeneic Hematopoietic Stem Cell Transplantation for Patients with Relapsed, High-Risk Multiple Myeloma. Biol Blood Marrow Transplant. 2016;22:258–267. doi: 10.1016/j.bbmt.2015.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tricot G, Vesole DH, Jagannath S, Hilton J, Munshi N, Barlogie B. Graft-versus-myeloma effect: proof of principle. Blood. 1996;87:1196–1198. [PubMed] [Google Scholar]
- 15.Noonan K, Borrello I. The immune microenvironment of myeloma. Cancer Microenviron. 2011;4:313–323. doi: 10.1007/s12307-011-0086-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gorgun GT, Whitehill G, Anderson JL, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013;121:2975–2987. doi: 10.1182/blood-2012-08-448548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.An G, Acharya C, Feng X, et al. Osteoclasts promote immune suppressive microenvironment in multiple myeloma: therapeutic implication. Blood. 2016;128:1590–1603. doi: 10.1182/blood-2016-03-707547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Goodyear O, Piper K, Khan N, et al. CD8+ T cells specific for cancer germline gene antigens are found in many patients with multiple myeloma, and their frequency correlates with disease burden. Blood. 2005;106:4217–4224. doi: 10.1182/blood-2005-02-0563. [DOI] [PubMed] [Google Scholar]
- 19.Brown RD, Spencer A, Ho PJ, et al. Prognostically significant cytotoxic T cell clones are stimulated after thalidomide therapy in patients with multiple myeloma. Leuk Lymphoma. 2009;50:1860–1864. doi: 10.3109/10428190903216804. [DOI] [PubMed] [Google Scholar]
- 20.Tyler EM, Jungbluth AA, O'Reilly RJ, Koehne G. WT1-specific T-cell responses in high-risk multiple myeloma patients undergoing allogeneic T cell-depleted hematopoietic stem cell transplantation and donor lymphocyte infusions. Blood. 2013;121:308–317. doi: 10.1182/blood-2012-06-435040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zeiser R, Bertz H, Spyridonidis A, Houet L, Finke J. Donor lymphocyte infusions for multiple myeloma: clinical results and novel perspectives. Bone Marrow Transplant. 2004;34:923–928. doi: 10.1038/sj.bmt.1704670. [DOI] [PubMed] [Google Scholar]
- 22.Orsini E, Alyea EP, Chillemi A, et al. Conversion to full donor chimerism following donor lymphocyte infusion is associated with disease response in patients with multiple myeloma. Biol Blood Marrow Transplant. 2000;6:375–386. doi: 10.1016/s1083-8791(00)70014-0. [DOI] [PubMed] [Google Scholar]
- 23.Peggs KS, Thomson K, Hart DP, et al. Dose-escalated donor lymphocyte infusions following reduced intensity transplantation: toxicity, chimerism, and disease responses. Blood. 2004;103:1548–1556. doi: 10.1182/blood-2003-05-1513. [DOI] [PubMed] [Google Scholar]
- 24.Montefusco V, Spina F, Patriarca F, et al. Bortezomib plus dexamethasone followed by escalating donor lymphocyte infusions for patients with multiple myeloma relapsing or progressing after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2013;19:424–428. doi: 10.1016/j.bbmt.2012.10.032. [DOI] [PubMed] [Google Scholar]
- 25.El-Cheikh J, Crocchiolo R, Furst S, et al. Lenalidomide plus donor-lymphocytes infusion after allogeneic stem-cell transplantation with reduced-intensity conditioning in patients with high-risk multiple myeloma. Exp Hematol. 2012;40:521–527. doi: 10.1016/j.exphem.2012.02.009. [DOI] [PubMed] [Google Scholar]
- 26.Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8:299–308. doi: 10.1038/nrc2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dudley ME, Yang JC, Sherry R, et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26:5233–5239. doi: 10.1200/JCO.2008.16.5449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Borrello I, Noonan KA. Marrow-Infiltrating Lymphocytes - Role in Biology and Cancer Therapy. Front Immunol. 2016;7:112. doi: 10.3389/fimmu.2016.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Noonan KA, Huff CA, Davis J, et al. Adoptive transfer of activated marrow-infiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma. Sci Transl Med. 2015;7:288ra278. doi: 10.1126/scitranslmed.aaa7014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cameron BJ, Gerry AB, Dukes J, et al. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 2013;5:197ra103. doi: 10.1126/scitranslmed.3006034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.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;122:863–871. doi: 10.1182/blood-2013-03-490565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gnjatic S, Nishikawa H, Jungbluth AA, et al. NY-ESO-1: review of an immunogenic tumor antigen. Adv Cancer Res. 2006;95:1–30. doi: 10.1016/S0065-230X(06)95001-5. [DOI] [PubMed] [Google Scholar]
- 33.Condomines M, Hose D, Raynaud P, et al. Cancer/testis genes in multiple myeloma: expression patterns and prognosis value determined by microarray analysis. J Immunol. 2007;178:3307–3315. doi: 10.4049/jimmunol.178.5.3307. [DOI] [PubMed] [Google Scholar]
- 34.Rapoport AP, Stadtmauer EA, Binder-Scholl GK, et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015;21:914–921. doi: 10.1038/nm.3910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sadelain M. T-cell engineering for cancer immunotherapy. Cancer J. 2009;15:451–455. doi: 10.1097/PPO.0b013e3181c51f37. [DOI] [PubMed] [Google Scholar]
- 36.Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3:388–398. doi: 10.1158/2159-8290.CD-12-0548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sadelain M. CAR therapy: the CD19 paradigm. J Clin Invest. 2015;125:3392–3400. doi: 10.1172/JCI80010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Geyer MB, Brentjens RJ. Review: Current clinical applications of chimeric antigen receptor (CAR) modified T cells. Cytotherapy. 2016;18:1393–1409. doi: 10.1016/j.jcyt.2016.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.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. 2015;385:517–528. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126:2123–2138. doi: 10.1172/JCI85309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.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;6:224ra225. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.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;5:177ra138. doi: 10.1126/scitranslmed.3005930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Garfall AL, Maus MV, Hwang WT, et al. Chimeric Antigen Receptor T Cells against CD19 for Multiple Myeloma. N Engl J Med. 2015;373:1040–1047. doi: 10.1056/NEJMoa1504542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Garfall AL, Stadtmauer EA, Maus MV, et al. Pilot Study of Anti-CD19 Chimeric Antigen Receptor T Cells (CTL019) in Conjunction with Salvage Autologous Stem Cell Transplantation for Advanced Multiple Myeloma. Am Soc Hematology. 2016 [Google Scholar]
- 46.Ramos CA, Savoldo B, Torrano V, et al. Clinical responses with T lymphocytes targeting malignancy-associated kappa light chains. J Clin Invest. 2016;126:2588–2596. doi: 10.1172/JCI86000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wijdenes J, Vooijs WC, Clement C, et al. A plasmocyte selective monoclonal antibody (B-B4) recognizes syndecan-1. Br J Haematol. 1996;94:318–323. doi: 10.1046/j.1365-2141.1996.d01-1811.x. [DOI] [PubMed] [Google Scholar]
- 48.Guo B, Chen M, Han Q, et al. CD138-directed adoptive immunotherapy of chimeric antigen receptor (CAR)-modified T cells for multiple myeloma. Journal of Cellular Immunotherapy. 2016;2:28–35. [Google Scholar]
- 49.Akl MR, Nagpal P, Ayoub NM, et al. Molecular and clinical profiles of syndecan-1 in solid and hematological cancer for prognosis and precision medicine. Oncotarget. 2015;6:28693–28715. doi: 10.18632/oncotarget.4981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Carpenter RO, Evbuomwan MO, Pittaluga S, et al. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res. 2013;19:2048–2060. doi: 10.1158/1078-0432.CCR-12-2422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Tai YT, Anderson KC. Targeting B-cell maturation antigen in multiple myeloma. Immunotherapy. 2015;7:1187–1199. doi: 10.2217/imt.15.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lee L, Bounds D, Paterson J, et al. Evaluation of B cell maturation antigen as a target for antibody drug conjugate mediated cytotoxicity in multiple myeloma. Br J Haematol. 2016;174:911–922. doi: 10.1111/bjh.14145. [DOI] [PubMed] [Google Scholar]
- 53.Dou H, Yan Z, Zhang M, Xu X. APRIL, BCMA and TACI proteins are abnormally expressed in non-small cell lung cancer. Oncol Lett. 2016;12:3351–3355. doi: 10.3892/ol.2016.5095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ali SA, Shi V, Maric I, et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood. 2016;128:1688–1700. doi: 10.1182/blood-2016-04-711903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cohen AD, Garfall AL, Stadtmauer EA, et al. B-Cell Maturation Antigen (BCMA)-Specific Chimeric Antigen Receptor T Cells (CART-BCMA) for Multiple Myeloma (MM): Initial Safety and Efficacy from a Phase I Study. Am Soc Hematology. 2016 [Google Scholar]
- 56.Turtle CJ, Hanafi LA, Berger C, et al. Immunotherapy of non-Hodgkin's lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8:355ra116. doi: 10.1126/scitranslmed.aaf8621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhang W, Zhao W, Liu J, et al. Phase I, open-label trial of anti-BCMA chimeric antigen receptor T cells in patients with relapsed/refractory multiple myeloma. Ferrata Storti Foundation via Giuseppe Belli 4, 27100; Pavia, Italy: 2017. pp. 2–3. [Google Scholar]
- 58.Mehta K, Shahid U, Malavasi F. Human CD38, a cell-surface protein with multiple functions. FASEB J. 1996;10:1408–1417. doi: 10.1096/fasebj.10.12.8903511. [DOI] [PubMed] [Google Scholar]
- 59.Atanackovic D, Steinbach M, Radhakrishnan SV, Luetkens T. Immunotherapies targeting CD38 in Multiple Myeloma. Oncoimmunology. 2016;5:e1217374. doi: 10.1080/2162402X.2016.1217374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Touzeau C, Moreau P. Daratumumab for the treatment of multiple myeloma. Expert Opin Biol Ther. 2017 doi: 10.1080/14712598.2017.1322578. [DOI] [PubMed] [Google Scholar]
- 61.Mihara K, Bhattacharyya J, Kitanaka A, et al. T-cell immunotherapy with a chimeric receptor against CD38 is effective in eliminating myeloma cells. Leukemia. 2012;26:365–367. doi: 10.1038/leu.2011.205. [DOI] [PubMed] [Google Scholar]
- 62.Drent E, Groen RW, Noort WA, et al. Pre-clinical evaluation of CD38 chimeric antigen receptor engineered T cells for the treatment of multiple myeloma. Haematologica. 2016;101:616–625. doi: 10.3324/haematol.2015.137620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tembhare P, Yuan C, Korde N, Maric I, Landgren O. Antigenic drift in relapsed extramedullary multiple myeloma: plasma cells without CD38 expression. Leuk Lymphoma. 2012;53:721–724. doi: 10.3109/10428194.2011.623257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Munoz P, Navarro MD, Pavon EJ, et al. CD38 signaling in T cells is initiated within a subset of membrane rafts containing Lck and the CD3-zeta subunit of the T cell antigen receptor. J Biol Chem. 2003;278:50791–50802. doi: 10.1074/jbc.M308034200. [DOI] [PubMed] [Google Scholar]
- 65.Kramer G, Steiner G, Fodinger D, et al. High expression of a CD38-like molecule in normal prostatic epithelium and its differential loss in benign and malignant disease. J Urol. 1995;154:1636–1641. [PubMed] [Google Scholar]
- 66.Casucci M, Nicolis di Robilant B, Falcone L, et al. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood. 2013;122:3461–3472. doi: 10.1182/blood-2013-04-493361. [DOI] [PubMed] [Google Scholar]
- 67.Shaffer DR, Savoldo B, Yi Z, et al. T cells redirected against CD70 for the immunotherapy of CD70-positive malignancies. Blood. 2011;117:4304–4314. doi: 10.1182/blood-2010-04-278218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hsi ED, Steinle R, Balasa B, et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin Cancer Res. 2008;14:2775–2784. doi: 10.1158/1078-0432.CCR-07-4246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Chu J, Deng Y, Benson DM, et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia. 2014;28:917–927. doi: 10.1038/leu.2013.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ghosh A, Holland AM, van den Brink MR. Genetically engineered donor T cells to optimize graft-versus-tumor effects across MHC barriers. Immunol Rev. 2014;257:226–236. doi: 10.1111/imr.12142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ghosh A, Koestner W, Hapke M, et al. Donor T cells primed on leukemia lysate-pulsed recipient APCs mediate strong graft-versus-leukemia effects across MHC barriers in full chimeras. Blood. 2009;113:4440–4448. doi: 10.1182/blood-2008-09-181677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ghosh A, Holland AM, Dogan Y, et al. PLZF confers effector functions to donor T cells that preserve graft-versus-tumor effects while attenuating GVHD. Cancer Res. 2013;73:4687–4696. doi: 10.1158/0008-5472.CAN-12-4699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ghosh A, Dogan Y, Moroz M, et al. Adoptively transferred TRAIL+ T cells suppress GVHD and augment antitumor activity. J Clin Invest. 2013;123:2654–2662. doi: 10.1172/JCI66301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ghosh A, Smith M, James SE, et al. Donor CD19 CAR T cells exert potent graft-versus-lymphoma activity with diminished graft-versus-host activity. Nat Med. 2017;23:242–249. doi: 10.1038/nm.4258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Brudno JN, Somerville RP, Shi V, et al. Allogeneic T Cells That Express an Anti-CD19 Chimeric Antigen Receptor Induce Remissions of B-Cell Malignancies That Progress After Allogeneic Hematopoietic Stem-Cell Transplantation Without Causing Graft-Versus-Host Disease. J Clin Oncol. 2016;34:1112–1121. doi: 10.1200/JCO.2015.64.5929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kochenderfer JN, Dudley ME, Carpenter RO, et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. 2013;122:4129–4139. doi: 10.1182/blood-2013-08-519413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Holland AM, Zakrzewski JL, Goldberg GL, Ghosh A, van den Brink MR. Adoptive precursor cell therapy to enhance immune reconstitution after hematopoietic stem cell transplantation in mouse and man. Semin Immunopathol. 2008;30:479–487. doi: 10.1007/s00281-008-0138-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zakrzewski JL, Suh D, Markley JC, et al. Tumor immunotherapy across MHC barriers using allogeneic T-cell precursors. Nat Biotechnol. 2008;26:453–461. doi: 10.1038/nbt1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hubner J, Hoseini SS, Suerth JD, et al. Generation of Genetically Engineered Precursor T-Cells From Human Umbilical Cord Blood Using an Optimized Alpharetroviral Vector Platform. Mol Ther. 2016;24:1216–1226. doi: 10.1038/mt.2016.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Pegram HJ, Purdon TJ, van Leeuwen DG, et al. IL-12-secreting CD19-targeted cord blood-derived T cells for the immunotherapy of B-cell acute lymphoblastic leukemia. Leukemia. 2015;29:415–422. doi: 10.1038/leu.2014.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Chung DJ, Pronschinske KB, Shyer JA, et al. T-cell Exhaustion in Multiple Myeloma Relapse after Autotransplant: Optimal Timing of Immunotherapy. Cancer Immunol Res. 2016;4:61–71. doi: 10.1158/2326-6066.CIR-15-0055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.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;110:296–304. doi: 10.1182/blood-2006-10-051482. [DOI] [PubMed] [Google Scholar]
- 83.Yousef S, Marvin J, Steinbach M, et al. Immunomodulatory molecule PD-L1 is expressed on malignant plasma cells and myeloma-propagating pre-plasma cells in the bone marrow of multiple myeloma patients. Blood Cancer J. 2015;5:e285. doi: 10.1038/bcj.2015.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Tamura H, Ishibashi M, Yamashita T, et al. Marrow stromal cells induce B7-H1 expression on myeloma cells, generating aggressive characteristics in multiple myeloma. Leukemia. 2013;27:464–472. doi: 10.1038/leu.2012.213. [DOI] [PubMed] [Google Scholar]
- 85.Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J Clin Oncol. 2016;34:2698–2704. doi: 10.1200/JCO.2015.65.9789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Berger R, Rotem-Yehudar R, Slama G, et al. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res. 2008;14:3044–3051. doi: 10.1158/1078-0432.CCR-07-4079. [DOI] [PubMed] [Google Scholar]
- 87.San Miguel J, Mateos MV, Shah JJ, et al. Pembrolizumab in Combination with Lenalidomide and Low-Dose Dexamethasone for Relapsed/Refractory Multiple Myeloma (RRMM): Keynote-023. Blood. 2015;126:505–505. [Google Scholar]
- 88.Lonial S, Ribeiro de Oliveira M, Yimer H, et al. 51TiPKEYNOTE-185: A randomized, open-label phase 3 study of pembrolizumab in combination with lenalidomide and low-dose dexamethasone in newly diagnosed and treatment-naive multiple myeloma (MM) Annals of Oncology. 2016;27 [Google Scholar]
- 89.John LB, Devaud C, Duong CP, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res. 2013;19:5636–5646. doi: 10.1158/1078-0432.CCR-13-0458. [DOI] [PubMed] [Google Scholar]
- 90.Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y. Multiplex Genome Editing to Generate Universal CAR T Cells Resistant to PD1 Inhibition. Clin Cancer Res. 2016 doi: 10.1158/1078-0432.CCR-16-1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Frassanito MA, Cusmai A, Dammacco F. Deregulated cytokine network and defective Th1 immune response in multiple myeloma. Clin Exp Immunol. 2001;125:190–197. doi: 10.1046/j.1365-2249.2001.01582.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Guillerey C, Nakamura K, Vuckovic S, Hill GR, Smyth MJ. Immune responses in multiple myeloma: role of the natural immune surveillance and potential of immunotherapies. Cell Mol Life Sci. 2016;73:1569–1589. doi: 10.1007/s00018-016-2135-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yeku OO, Brentjens RJ. Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy. Biochem Soc Trans. 2016;44:412–418. doi: 10.1042/BST20150291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016;13:394. doi: 10.1038/nrclinonc.2016.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Pegram HJ, Lee JC, Hayman EG, et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012;119:4133–4141. doi: 10.1182/blood-2011-12-400044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hsu C, Hughes MS, Zheng Z, Bray RB, Rosenberg SA, Morgan RA. Primary human T lymphocytes engineered with a codon-optimized IL-15 gene resist cytokine withdrawal-induced apoptosis and persist long-term in the absence of exogenous cytokine. J Immunol. 2005;175:7226–7234. doi: 10.4049/jimmunol.175.11.7226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Singh H, Figliola MJ, Dawson MJ, et al. Reprogramming CD19-specific T cells with IL-21 signaling can improve adoptive immunotherapy of B-lineage malignancies. Cancer Res. 2011;71:3516–3527. doi: 10.1158/0008-5472.CAN-10-3843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hsu C, Jones SA, Cohen CJ, et al. Cytokine-independent growth and clonal expansion of a primary human CD8+ T-cell clone following retroviral transduction with the IL-15 gene. Blood. 2007;109:5168–5177. doi: 10.1182/blood-2006-06-029173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Cherkassky L, Morello A, Villena-Vargas J, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016;126:3130–3144. doi: 10.1172/JCI83092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Zhao Z, Condomines M, van der Stegen SJ, et al. Structural Design of Engineered Costimulation Determines Tumor Rejection Kinetics and Persistence of CAR T Cells. Cancer Cell. 2015;28:415–428. doi: 10.1016/j.ccell.2015.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Long AH, Haso WM, Shern JF, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015;21:581–590. doi: 10.1038/nm.3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Curran KJ, Seinstra BA, Nikhamin Y, et al. Enhancing antitumor efficacy of chimeric antigen receptor T cells through constitutive CD40L expression. Mol Ther. 2015;23:769–778. doi: 10.1038/mt.2015.4. [DOI] [PMC free article] [PubMed] [Google Scholar]