Immunotherapeutic Strategies for Multiple Myeloma


Mechanism

Ag

Short description of function

Ab name

Results of earlier trials

Ongoing clinical trials

mAbs targeting proteins involved in myeloma cell adhesion

CS1

See text

Elotuzumab
  
Syndecan-1 (CD138)

Cell surface heparan sulfate proteoglycan. Receptor for the extracellular matrix molecules collagen and fibronectin, also binds via its heparan sulfate chains to various soluble extracellular molecules, including hepatocyte growth factor and fibroblast growth factor [26, 127]

nBT062
 
A phase I/IIa multidose-escalation study of BT062 in combination with lenalidomide and dexamethasone in subjects with relapsed or relapsed/refractory multiple myeloma (NCT01638936)
  
nBT062-SPBD-DM4

In a phase I study of BT062 conjugated to DM4 in RRMM, 13 of 32 patients (40 %) achieved stable disease [128]

A phase I/IIa multidose-escalation study to evaluate maximum tolerated dose (MTD), pharmacokinetics (PK), safety and efficacy of BT062 in subjects with relapsed or relapsed/refractory multiple myeloma (NCT01001442)

CD56

Membrane glycoprotein from the immunoglobulin superfamily expressed on muscle cells and neurons. It appears to mediate cell adhesion, migration, invasion, and anti-apoptosis

Lorvotuzumab (IMGN901)

In a phase I lorvotuzumab monotherapy study in heavily pretreated MM patients OR was 18 % (n = 28) with 2 PR and 3 MR [129]
    
In a phase I combination study of lorvotuzumab with lenalidomide and dexamethasone in RRMM, the ORR was 59 % (n = 32) with 1 sCR, 1 CR, 8 VGPR, and 9 PR. Higher doses complicated by increased incidence of peripheral neuropathy [130]

NCT00991562 mentioned to the left

CD38

See text

Daratumumab
  
Intercellular adhesion molecule-1 (ICAM-1) (CD54)

ICAM-1 mediates adhesion of myeloma cells to bone marrow stromal cells and thereby contributes to cell adhesion-mediated drug resistance

BI-505
 
A phase I dose-escalation study to determine the safety, pharmacokinetics, and pharmacodynamics of BI-505, in patients with relapsed/refractory multiple myeloma (NCT01025206)
    
A single-arm, open-label, phase II clinical trial evaluating disease response following treatment with BI-505, in patients with smoldering multiple myeloma (NCT01838369)

mAbs neutralizing growth factors or inhibiting growth-promoting receptors

IL-6 and IL-6 receptor

See text

Siltuximab; CNTO 328
  
mAb 1339; hPM1; tocilizumab

Insulin-like growth factor-1 receptor (IGF-1)

IGF-1 is an important growth factor for myeloma cells. It acts synergistically with IL-6 on myeloma cell growth and survival. IGF-1 also protects myeloma cells against anticancer drugs including dexamethasone and bortezomib [41, 131, 132]. IGF-1R is aberrantly expressed on MM cells in about 75 % of the cases and is associated with disease severity [132, 133]

AVE1642

In a phase I study of the anti-(IGF-1R)mAb, AVE1642, as single agent and in combination with bortezomib in patients with relapsed MM, in the monotherapy arm, the drug was well tolerated with no MTD reached (1 DLT of hyperglycemia). Of 15 patients, 1 patient had MR and 7 SD. In the combination arm, out of 11 patients only 2 (18 %) had objective response (1 CR and 1 PR) which was considered insufficient [134]
   
Figitumumab (CP-751,871)

In a phase I study of figitumumab in RRMM, no DLT were identified. Monotherapy did not result in objective responses. Dexamethasone could be added when less than PR was achieved. Of the 27 patients with the combination therapy 6 achieved PR and 3 MR [135]
   
IMC-A12
  
Vascular endothelial growth factor (VEGF)

Key cytokine that promotes angiogenesis in a variety of tumor types including in the bone marrow and therefore contributing to myeloma growth and survival. VEGF also functions as a growth factor for myeloma cells, stimulates IL-6 production in stromal cells, triggers myeloma cell migration, upregulates antiapoptotic proteins, and inhibits maturation of dendritic cells [26, 41]

Bevacizumab

In a phase II randomized trial of bevacizumab versus bevacizumab and thalidomide for RRMM, in the bevacizumab alone arm, 1 patient achieved SD (n = 6) with median EFS of only 49d for the cohort. For the combination arm, 2 PR and 3 SD (n = 6), similar results as single-agent thalidomide [136]

Bevacizumab, lenalidomide, and dexamethasone in treating patients with relapsed or refractory stage II or stage III multiple myeloma (NCT00410605)
   
In a randomized phase II study of bevacizumab and bortezomib versus bortezomib in RRMM (AMBER), the ORR was similar between the two arms, 51 % in the combination arm (n = 49) and 43.4 % in the bortezomib monotherapy arm (n = 53). The median response duration was 6.9 m and 6 m, respectively [137]
     
In a phase II trial of bevacizumab, lenalidomide, and dexamethasone in RRMM, the addition of bevacizumab did not translate to either increased response rate or prolongation of PFS (ORR 64 %, n = 36) with some increased gastrointestinal and cardiac toxicity [138]

A phase II trial of bevacizumab combined with lenalidomide and dexamethasone (BEV/REV/DEX) in relapsed or refractory multiple myeloma (NCT00410605)
   
A phase II study of bevacizumab and bortezomib in patients with RRMM (NCT00464178)
 
B-cell-activating factor (BAFF)

BAFF is a member of the tumor necrosis factor superfamily. It is produced in the bone marrow microenvironment by monocytes, osteoclasts, and neutrophils [139]. BAFF triggers activation of NF-κB, PI3K, and MAPK pathways, resulting in survival and dexamethasone resistance of myeloma cells [140]. BAFF also increases adhesion of MM cells to BMSCs [141]

Tabalumab (LY2127399)
 
A multicenter, randomized, double-blind, placebo-controlled phase II study of tabalumab in combination with bortezomib and dexamethasone in patients with previously treated multiple myeloma (NCT01602224)
    
A phase I study of LY2127399 in combination with bortezomib and dexamethasone in Japanese patients with relapsed or refractory multiple myeloma NCT01556438
    
A phase I safety study of LY2127399 in combination with bortezomib in patients with relapsed or refractory multiple myeloma (NCT00689507)

CD74

CD74 is a transmembrane protein that forms the invariant portion of HLA-DR. CD74 also functions as a receptor for macrophage migration-inhibitory factor [142]. Its activation leads to proliferation and survival of cells through activation of signaling cascades including the NF-kB pathway [143]

Milatuzumab (hLL1, IMMU-115),

In a phase I dose-escalating trial of milatuzumab in RRMM, 2 DLT (infusion reaction, unexplained anemia) and 5 SAEs (bacterial meningitis, confusion/hypercalcemia, fever, cord compression, epistaxis/thrombocytopenia) were reported. No OR observed. 4 of 21 patients had disease stabilization for at least 3 months [144]
   
IMMU-110-doxorubicin conjugated to milatuzumab
 
A phase I/II study of hLL1-DOX (milatuzumab-doxorubicin antibody-drug conjugate) in patients with multiple myeloma (NCT01101594)
 
CD40

CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily and is highly expressed on MM cells and on BMSCs. CD40 activation induces an array of biological effects, including MM cell proliferation and migration. In addition, CD40 stimulation is important for myeloma cells adhesion to BMSCs, leading to augmented production of IL-6 and VEGF in these cells [145148]

Dacetuzumab (SGN-40)

In a phase I multidose study of dacetuzumab in advanced MM patients, the maximum tolerated dose was 12 mg/kg/week. 9 of 44 patients (20 %) had a best clinical response of stable disease [149]
    
In a phase Ib, dose-escalation study of dacetuzumab, lenalidomide, and low-dose dexamethasone, the combination was well tolerated. 13 of 33 evaluable patients (39 %) achieved an objective response (1 CR, 12 PR); other responses were 4 MR, 10 SD [150]
   
XmAbCD40
    
Lucatumumab HCD122; CHIR-12.12

In an open-label, multicenter, phase I study of lucatumumab in RRMM, the MTD was 4.5 mg/kg. 12 of 28 patients (43 %) had SD and 1 PR [151]
 
mAbs-activating death receptors

TRAIL-R1 and TRAIL-R2

TNF-related apoptosis-inducing ligand (TRAIL) is a member of the death receptor ligand family, a subclass of the tumor necrosis factor family. Its binding induces formation of a death-inducing signaling complex, ultimately leading to caspase activation and initiation of apoptosis. The physiological function of TRAIL is reported to be in immune surveillance and immune-mediated tumor suppression [152]

Mapatumumab, HGS-ETR1

In a phase II trial of mapatumumab in combination with bortezomib in patients with bortezomib naïve RRMM, the addition of mapatumumab to bortezomib did not increase ORR, PFS, or duration of response compared to bortezomib alone. ORR was 51 % for bortezomib alone versus 42 % in the combination arm (n = 104) [153]
   
Lexatumumab, HGS-ETR2
  
mAbs improving the antitumor immune response

Killer-cell immunoglobulin-like receptors (KIR)

KIRs are transmembrane glycoproteins expressed by natural killer cells and subsets of T cells and function as key regulators of NK cell activity [154]

1–7F9/IPH2101

In a phase I trial of the anti-KIR antibody IPH2101 in patients with RRMM, the drug was safe and tolerable. No objective responses were observed. 11 of 32 patients (34 %) achieved a best response of stable disease [117]

Multicenter phase II study on the antitumor activity, safety, and pharmacology of two dose regimens of IPH2101, in patients with smoldering multiple myeloma (KIRMONO) (NCT01222286)
   
In interim results of a phase I trial of the anti-inhibitory KIR antibody, IPH2101, and lenalidomide in MM of 13 patients, 1 achieved unconfirmed CR, 3 PR, 2 MR, and 1 SD [155]

Multicenter phase I study on the safety, antitumor activity, and pharmacology of IPH2101, a human monoclonal anti-KIR, combined with lenalidomide in patients with multiple myeloma experiencing a first or second relapse (NCT01217203)
    
Randomized phase II study evaluating the antitumor activity, safety, and pharmacology of two dose regimens of IPH2101, in patients with multiple myeloma in stable partial response after a first-line therapy (NCT00999830)
    
A phase II trial of IPH2101 (anti-KIR) in smoldering multiple myeloma (NCT01248455)

PD-L1

See text

CT-011
  
CD200

D200 is a highly conserved type I transmembrane glycoprotein that is expressed on many cell types. The expression of the receptor for CD200 (CD200R1) is restricted to myeloid-derived antigen-presenting cells and certain populations of T cells. CD200 imparts an immunoregulatory signal through CD200R, leading to the suppression of T-cell-mediated immune responses. CD200 is expressed on MM cells of the majority of newly diagnosed MM patients. And its expression predicts poor prognosis for patients receiving ASCT [156]

Samalizumab (ALXN6000)

In a phase I/II study, single-agent samalizumab was evaluated in patients with myeloma or chronic lymphocytic leukemia (n = 26; 3MM). Treatment was well tolerated with antitumor activity evident in some patients receiving multiple cycles, which was associated with reduction in regulatory T cells and increase in activated T cells [157]
 
mAbs targeting mediators of bone disease

RANKL

Skeletal effects of MM believed to be mediated through myeloma-derived cytokines which have been demonstrated to induce RANKL production. RANKL promotes the proliferation, differentiation, survival, and fusion of osteoclastic precursor cells, activates osteoclastic precursor cells toward mature osteoclasts, and inhibits apoptosis of mature osteoclasts. RANKL exerts its biologic effects through binding to and activating the specific receptor, RANK, a transmembrane member of the TNFR superfamily [158]

Denosumab

In a phase II trial of denosumab in the treatment of relapsed or plateau-phase MM, denosumab effectively inhibited the RANKL pathway as evidenced by suppressed levels of the bone turnover marker, serum C-terminal telopeptide of type 1 collagen. However, no patient had an objective response [159]

A randomized, double-blind, multicenter study of denosumab compared with zoledronic acid (Zometa®) in the treatment of bone disease in subjects with newly diagnosed multiple myeloma (NCT01345019)
    
In a phase III trial, denosumab was noninferior (trending to superiority) to zoledronic acid (2A) in preventing or delaying time to first on-study skeletal-related events (SRE) in patients with advanced cancer metastatic to bone or myeloma (number of myeloma patients, 180 of 1,776 included in the study) [160]

An open-label, single-arm, extension study to evaluate the long-term safety of denosumab in the treatment of bone metastases in subjects with advanced cancer or multiple myeloma (NCT00950911)
 
Dickkopf-1

Dickkopf-related protein 1 (DKK1), a soluble Wnt-signaling inhibitor, is produced by myeloma cells. It inhibits osteoblast differentiation, and its expression is correlated with the presence of lytic bone lesions in patients with MM [161, 162]

BHQ880

In a phase I/II study of BHQ880 in combination with ZA in patients with RRMM with prior SRE, the combination treatment was well tolerated [163]

A single-arm, open-label, phase II clinical trial evaluating disease response following treatment with intravenous BHQ880, in previously untreated patients with high-risk, smoldering multiple myeloma (NCT01302886)
    
In a phase II study in previously untreated patients with smoldering MM at risk for progression, BHQ880 given as monotherapy was well tolerated and resulted in the first evidence of anabolic bone activity using a novel imaging modality (increases in bone strength at 6 months, in 4 of 5 patients evaluated using quantitative computed tomography with finite element analysis) [164]
 





4.4 Cellular Immunotherapy for Multiple Myeloma



4.4.1 Allogeneic Transplantation


The unique potential efficacy of cellular immunotherapy for myeloma is highlighted by the observation that allogeneic transplantation induces durable remissions in a subset of patients due to the graft-versus-myeloma effect [5053]. A summary of early data of myeloablative transplantation from the European Bone Marrow Transplant Registry demonstrated that 28 % of patients remained in remission 7 years posttransplant, suggesting that durable responses were potentially achievable [54]. However, the median survival was only 10 months due to extremely high treatment-related mortality, raising a difficult choice for physicians and patients as to the applicability of this strategy. In a more recent report of 158 patients undergoing autologous or allogeneic transplantation based on donor availability, the event-free survival (EFS) following allogeneic transplantation was 33 and 31 % at 5 and 10 years, respectively, consistent with the presence of a subgroup that appear to have sustained disease response [55]. The role of the graft-versus-myeloma effect in preventing disease recurrence was further supported by a retrospective report from the European registry, in which patients with limited or extensive chronic graft-versus-host (cGVHD) disease demonstrated markedly improved 3-year survival (84 and 58 %, respectively) as compared to those without cGVHD (29 %) [56].

Donor lymphocyte infusion (DLI) as a treatment for posttransplant relapse has been shown to induce disease response, achievement of molecular remission, reconstitution of TCR Vβ repertoire, and long-term disease control in a subset of patients [5760]. However, DLI therapy is complicated by GVHD due to the lack of myeloma specificity of the alloreactive lymphocytes [61]. Efforts to limit toxicity through the use of reduced intensity conditioning regimens have resulted in a decrease in treatment-related mortality but a concomitant increase in the risk of relapse. As such, immune-based targeting of myeloma cells by alloreactive lymphocytes may carry the unique potential for curative outcomes; nonetheless, the lack of specificity and toxicity significantly limits its use.

Investigators have examined strategies to induce autologous cellular immune responses that selectively target myeloma-associated antigens while minimizing toxicity to normal tissue.

One such strategy is the use of cancer vaccines to foster the expansion of tumor-specific lymphocytes. Myeloma-associated antigens that have been explored as targets for immunotherapy include the idiotype protein, MUC1, WT1, PRAME, CYP1B1, and HSP96 [6269]. Vaccine strategies have included the introduction of myeloma-specific antigens in the context of immune adjuvants and the loading of individual or whole-cell-derived antigens onto antigen-presenting cells such as DCs.


4.4.2 DC-Based Vaccines as a Platform for Antigen Presentation


DCs represent a diverse network of antigen-presenting cells that play a prominent role in mediating immune responsiveness [70]. Circulating DC populations have been identified as myeloid and plasmacytoid in origin with the capacity to elicit Th1 and Th2 responses, respectively. Plasmacytoid DCs have been shown to contribute to the stromal environment in myeloma and may contribute to tumor-mediated tolerance [4]. Myeloma antigens administered in the context of immune adjuvants may recruit and activate native DC populations that subsequently internalize and present tumor antigens [7173]. However, functional deficiencies have been demonstrated in DCs derived from myeloma patients which may impact their ability to elicit immunologic responses [5]. Alternatively, myeloid DCs with strong expression of costimulatory molecules and stimulatory cytokines may be generated ex vivo through cytokine stimulation of precursor populations [74]. DCs generated ex vivo and loaded with myeloma-associated antigens may act as a platform for cancer vaccines [75]. Strategies to introduce tumor antigens include pulsing with peptides, proteins, or lysates [76], electroporation with tumor-derived RNA or DNA [8, 77, 78], loading of tumor-derived apoptotic bodies [79], transduction with viral vectors expressing tumor antigens potentially enhanced by costimulatory molecules [8083], and the use of whole-cell fusion between DCs and myeloma cells [8486].


4.4.3 Myeloma Vaccines: Single-Antigen Approaches


The idiotype protein represents a truly tumor-specific antigen created by the unique immunoglobulin gene arrangement intrinsic to the malignant clone [87]. Vaccination with the idiotype protein in conjunction with granulocyte-macrophage colony-stimulating factor (GM-CSF) or IL-12 was associated with antigen-specific T-cell responses. Prolonged disease-free progression was observed in patients exhibiting an immunologic response [88]. Responses have also been observed following vaccination with antigen-presenting cells pulsed with M protein or with DCs loaded with idiotype and exposed to CD40L to induce maturation [8993]. Vaccination with idiotype-pulsed antigen-presenting cells posttransplant was associated with improved progression-free survival as compared to a historical control cohort.

A peptide-based vaccine for WT1 administered with immune adjuvant has been shown to elicit immunologic response in patients with hematological malignancies and a decrease in measures of disease [94, 95]. In a recent study, WT1-specific immunity following allogeneic transplantation for myeloma was associated with long-term disease control. Peptide-based vaccine for MUC1 is currently being explored in patients with myeloma (NCT01232712). Expression of several cancer-testis antigens has been demonstrated and has been shown to be targeted by donor-derived humoral responses following allogeneic transplantation, confirming their potential immunogenicity. The cancer-testis antigen, NY-ESO, demonstrates increased expression by plasma cells in the setting of advanced disease, creating an appealing target for immune-based therapy [96]. Repetitive stimulation with DCs pulsed with an NY-ESO-derived peptide elicits a strong CTL response in vitro, demonstrating an activated phenotype capable of lysing primary myeloma cells [97]. Recent studies have identified a series of antigens recognized by T cells in patients following syngeneic transplantation.

Several other peptides which are highly expressed on myeloma cells and are important in the pathogenesis of the disease have been identified as potential immunogenic targets. Heteroclitic XBP1 (X-box-binding protein 1) (unspliced 184–192 and spliced 367–375), CD138 (syndecan-1)260–268, and CS1239-247 were shown each alone and in a cocktail combination of the four to generate specific CTLs enriched for effector and activated T cells, Ag-specific cytotoxicity against MM cell lines, as well as increased degranulation, proliferation, and INF-γ secretion [98101].


4.4.4 Myeloma Vaccines: Whole-Cell Approaches


The use of whole-cell-derived antigens for vaccination may elicit a broad polyclonal response that is better able to target the heterogeneity of the myeloma cell population [86]. Consistent with this hypothesis, a murine model demonstrated the emergence of idiotype-negative variants following idiotype-based vaccinations, while whole myeloma cell-based vaccines did not induce resistance [102]. DCs pulsed with tumor lysates have been shown to induce myeloma-associated immunity, although the clinical efficacy was uncertain [76].

The authors have developed a vaccine model in which patient-derived myeloma cells are fused with autologous DCs, creating a hybridoma which expresses a broad array of myeloma antigens in the context of enhanced costimulation [86]. In a murine model, DC/MM fusions were shown to be protective against lethal challenge with syngeneic myeloma cells, and therapeutic efficacy was further enhanced by coadministration of IL-12 [103]. In preclinical human studies, fusion of DCs and MM cells elicited the expansion of activated T cells that potently lysed autologous myeloma cells in vitro.

A phase I clinical trial was completed in which successive cohorts of patients with advanced myeloma underwent vaccination with escalating doses of autologous DC/MM fusions [104]. Patients had undergone a median of four prior treatment regimens. Myeloma cells were derived from bone marrow aspirates, and DCs were generated from adherent mononuclear cells cultured with GM-CSF and IL-4 and matured with TNF-α. Patients underwent serial vaccination in conjunction with GM-CSF. Vaccine-associated toxicity consisted of transient grade 1–2 vaccine site reactions most commonly, while clinically significant autoimmunity was not observed. Biopsy of the vaccine bed demonstrated a dense infiltrate of CD8+ T cells consistent with T-cell expansion occurring at the site of vaccination. Vaccination resulted in the expansion of myeloma-specific T cells in the majority of patients as manifested by the percent of CD4+ and/or CD8+ T cells expressing IFN-γ following ex vivo exposure to autologous tumor lysate. On SEREX analysis, humoral responses against novel proteins were noted after vaccination. These findings were consistent with the induction of myeloma-specific immunity in patients with advanced disease. Of note, 66 % of patients demonstrated a period of disease stability ranging from several months to greater than 2 years after vaccination.

The authors have completed a phase II clinical trial in which patients underwent vaccination with DC/MM fusions in conjunction with autologous stem cell transplantation. It was postulated that vaccine response would be augmented following transplant-mediated cytoreduction and in the context of lymphopoietic reconstitution with the associated depletion of regulatory T cells. It was demonstrated that the posttransplant period was associated with the expansion of myeloma-reactive T cells which were further boosted by vaccination with DC/MM fusions. Vaccination was associated with the conversion of partial to complete responses greater than 100 days posttransplant in a subset of patients. A clinical trial is now underway examining the efficacy of PD-1 blockade in conjunction with the DC/MM fusion vaccine following autologous transplantation, and a national cooperative group study for the assessment of DC/MM fusion vaccine with lenalidomide versus lenalidomide maintenance alone is being planned.


4.4.5 NK Cell Therapy


Augmentation of NK cell-mediated immunity has been explored as therapy for MM. Preclinical models have demonstrated that thalidomide and lenalidomide increase the production of IL-2 by T cells, which stimulates NK cell activation and function against MM [105]. Lenalidomide has also been shown to increase CD16 and LFA-1 expression on NK cells, which facilitates an ADCC response against MM [106]. Lenalidomide also modulates the balance of NK cell-activating and inhibitory ligand expression on MM cells. It decreases expression of PD-L1 and enhances expression of ULBP-1(NKG2D ligand) on MM cells, which both result in improved NK cell immune response, as well as recognition and lysis of MM tumor targets [20, 107]. Bortezomib decreases MM expression of MHC class I and enhances the sensitivity of myeloma to NK cell-mediated lysis [108].

The importance of NK cell-mediated immunity in modulating disease outcome was highlighted by the observation that levels of autologous NK cells reinfused with autologous transplantation correlates with absolute lymphocyte recovery after ASCT for MM and non-hodgkin lymphoma [109]. Lymphocyte subset analyses revealed that an absolute NK cell count of 80/μL or more on day +15 post-SCT correlated significantly with improved progression-free survival [110]. In patients with MM undergoing allogeneic SCT, killer-cell immunoglobulin-like receptor (KIR)-ligand mismatch predicting for NK activation was protective against relapse [111]. In addition, the infusion of T-cell-depleted, haploidentical, KIR-mismatched NK cells, followed by delayed autograft stem cell rescue, has been shown to induce a near-complete/complete response rate of nearly 50 % [112]. Improved disease-free and overall survival was observed in myeloma patients who received grafts from donors with KIR haplotype B, which is associated with more activating receptor genes than KIR haplotype A [113]. Lenalidomide therapy for patients with progressive MM following allogeneic SCT has been associated with an overall response rate of 66 %, and immunomonitoring data show that lenalidomide augments NK cell expression of the activating receptor NKp44 [114]. Moreover, in a recent phase I/II study of lenalidomide given early after allogeneic SCT for MM, lenalidomide treatment resulted in an increase of activating receptors NKp30 and NKp44 on NK cells, as well as an increase in NK cell-mediated cytotoxicity directed against myeloma associated with an increase in the rate of complete remission [115].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Feb 18, 2017 | Posted by in ONCOLOGY | Comments Off on Immunotherapeutic Strategies for Multiple Myeloma

Full access? Get Clinical Tree

Get Clinical Tree app for offline access