Immunotherapies that are currently being studied in multiple  myeloma are discussed in this section under four headings:  checkpoint inhibitors, chimeric antigen receptor (CAR) T cells,  bispecific antibodies, and antibody drug conjugates (fig 2).
    Malignant plasma cells in most patients with multiple myeloma    express the checkpoint programmed death-ligand 1, which is    upregulated especially when exposed to inflammatory mediators    such as interferon . Interaction of this checkpoint molecule    with programmed cell death protein 1 on T cells limits their    proliferation and cytotoxic activity.6869  
    The first study evaluating single agent nivolumab for relapsed    multiple myeloma showed a response in only one of 27    patients.7071 Despite    a lack of single agent activity, single arm trials combining    checkpoint inhibitors with immunomodulatory imide drugs and    dexamethasone because of the potential synergy72 look promising.7374  
    With these clinical data, three large randomized phase III    trials were halted by the FDA in 2017 because of increased    serious adverse events and deaths as well as decreased overall    survival in the checkpoint inhibitor arm (pomalidomide and    dexamethasone with and without pembrolizumab) in relapsed and    refractory multiple myeloma (hazard ratio 1.61, 95% confidence    interval 0.91 to 2.85),75 pomalidomide and    dexamethasone with and without nivolumab in relapsed and    refractory multiple myeloma (1.19, 0.64 to 2.20),76 and lenalidomide and dexamethasone with    and without pembrolizumab in transplant ineligible patients    with newly diagnosed multiple myeloma (2.06, 0.93 to    4.55)77).  
    These trials encourage caution with expedited timelines for    future combination studies for drugs with limited single agent    activity.7879 Future    trials in multiple myeloma will need to be based on sound    preclinical and clinical rationale with other partners and be    conducted in heavily treated patients (with limited standard    options) initially.  
    CAR T cells are human T cells that have been genetically    modified and expanded in the laboratory before they are infused    back into patients to target the tumor. The receptor on the    surface of CAR T cells that targets the tumor antigens consists    of several parts (fig 3): an extracellular,    non-major histocompatibility complex restricted, targeting    domain, usually derived from a single chain variable fragment    of a monoclonal antibody; a spacer region; a transmembrane    domain; an intracellular signaling domain including the CD3    activation domain; and a costimulatory domain (eg, CD28 or    4-1BB). The single chain variable fragment was originally    derived from mice (hence the term chimeric), but many of the    newer constructs are fully human.8081  
            Chimeric antigen receptor (CAR) T cell structure          
    CD3 positive T cells are obtained from patients (for autologous    CAR T cells) or healthy donors (for allogeneic CAR T cells) via    a process called leukapheresis. These T cells are expanded    manifold in culture and activated using beads coated with    anti-CD3 or anti-CD28 monoclonal antibodies or cell based    artificial antigen presenting cells.82    The T cells are then transduced with a vector (usually either    lentiviral or retroviral) that carries the gene encoding a    receptor to an antigen present on the surface of tumor cells.    This manufacturing process takes up to four weeks at a good    manufacturing practices facility, and the CAR T cells can then    be stored until needed by the patient. This delay means that    the disease must not be rapidly progressing, so that the    patient is able to wait until the CAR T product is ready;    otherwise the patient will need bridging chemotherapy. Two to    seven days before CAR T cell infusion, a patient receives    lymphodepleting chemotherapy to make way for the CAR T cells    that are subsequently given as an intravenous infusion. Once    infused into patients, the CAR T cells encounter the antigen,    proliferate, and kill the tumor cells (fig 4).    These cells, therefore, combine the target specificity of a    monoclonal antibody with the enhanced cytotoxicity of T cells    without requiring human leucocyte antigen presentation of the    target antigen.83  
            Chimeric antigen receptor (CAR) T cell treatment for            multiple myelomasequence of events. CRS=cytokine            release syndrome; ICANS=immune effector cell associated            neurotoxicity syndrome          
    An ideal antigen is one that is widely and exclusively    expressed on cancer cells but not on normal cells to enhance    efficacy and reduce toxicity.8485 In multiple myeloma, most emerging    immunotherapies (including CAR T cells) target B cell    maturation antigen (BCMA), a type III transmembrane receptor,    which is a promising target antigen.8687 BCMA is also known    as tumor necrosis factor receptor superfamily member 17 or    CD269. It is expressed in nearly all plasma cells (normal and    malignant) although its expression is variable.88 BCMA promotes plasma cell survival and    is induced during plasma cell differentiation89 by binding to ligands (a proliferation    inducing ligand (APRIL) and B cell activating factor (BAFF))    that are produced by osteoclasts.90    Increased levels of soluble BCMA are associated with high tumor    burden in multiple myeloma and thus worse outcomes.91  
    CAR T cells targeting CD19 were approved by the FDA in 2017 for    refractory large B cell lymphoma9293 and acute lymphoblastic leukemia,94 and are being used in clinical practice.    The first study on CAR T cell treatment directed by BCMA opened    in 2014 at the US National Cancer Institute.879596    Since then, about a dozen of different early phase clinical    trials have been conducted on BCMA CAR T treatment for advanced    multiple myeloma.9798 A    detailed review outlining the differences in the construct,    manufacturing, and clinical efficacy of these different    products has been published previously.8186  
    This review focuses on four BCMA CAR T cell products that are    currently being evaluated in registration (that is, for    regulatory approval) phase I/II clinical trials for patients    with relapsed and refractory multiple myeloma. These products    include bb2121 (now known as idecabtagene vicleucel or    ide-cel), JCARH125 (now known as orvacabtagene autoleucel or    orva-cel), LCAR-B38M (now known as JNJ-4528), and P-Bcma-101.    FDA approvals for some of these agents are anticipated in    2020-21 for relapsed and refractory multiple myeloma (fig 5; table    2). The high overall response rates of 60-100% seen in    these trials in a highly refractory population is    unprecedented, although the durability of these responses is    still in question.  
            Four major constructs of chimeric antigen receptor            (CAR) T cells targeting B cell maturation antigens            (BCMA), currently in multicenter clinical trials            investigating multiple myeloma. This figure does not            include all BCMA constructs in multiple myeloma.            ScFv=single chain variable fragment; VH            only=variable-heavy chain only fragments          
          Summary of major multicenter clinical trials          investigating multiple myeloma treatments*        
      The most advanced CAR T cell treatment targeting a BCMA is      ide-cel (bb2121), which uses a lentiviral vector for CAR      insertion and includes a 4-1BB costimulatory domain as well      as a murine single chain variable fragment.114 In a phase I non-randomized, open      label, multicenter trial in relapsed and refractory multiple      myeloma (3 prior lines of treatment) for 33 patients treated      at various doses,115 researchers      found an overall response rate of 85% with a median      progression free survival of 11.8 months. A higher overall      response rate was seen at the higher dose levels and doses of      150-450106 CAR T cells were defined as the active      dose.115 This dose is being tested      currently in a multicenter, single arm, open label trial to      evaluate bb2121 CAR T cells further in relapsed and      refractory multiple myeloma; the trial has completed      enrolment of 149 patients worldwide. Preliminary results show      an overall response rate of 73% (complete response rate 33%)      and median progression free survival of 8.8 months in 128      patients treated at doses of 150-450106 cells      (table 2).99      Fifty four patients treated at the highest dose level of      450106 cells had an overall response rate of 82%      and a median progression free survival of 12.1 months.99 These results have been submitted to      regulatory agencies including the FDA and European Medicines      Agency for treatment for advanced multiple myeloma.    
      Orva-cel (JCARH125) is another second generation CAR product      with a fully human B cell derived single chain variable      fragment, a 4-1BB costimulatory domain, and optimized      manufacturing (predefined CD4:CD8 ratio) that is derived from      preclinical work at Memorial Sloan Kettering Cancer Center.      The preliminary data for the multicenter phase I/II EVOLVE      study were presented at the American Society of Clinical      Oncology meeting in 2020. These patients had received a      median of six prior treatments. They received escalating      doses of 50-600106 cells. The results for 62      patients treated at the 300-600106 cells dose      range showed an overall response rate of 92% (complete      response rate 36%).100101 The trial is currently enrolling at      the recommended phase II dose of 600106 cells      (table 2).    
      The LCAR-B38M CAR construct was developed initially in China      and is currently being pursued in the US and globally as      JNJ-4528 (table 2). It      consists of two llama derived variable-heavy chain only      fragments that target two epitopes of BCMA designed to confer      avidity. In a phase I/II study in China, researchers found      deep durable responses with a median progression free      survival of 19.9 months and a manageable safety profile in      relapsed and refractory multiple myeloma, although the      patients in this study were treated earlier in their disease      course with a median of three prior lines of treatment and      were therefore less heavily pre-treated.102103104105 In      the US and Europe, a multicenter phase Ib/II clinical trial      of this CAR construct as JNJ-4528 in relapsed and refractory      multiple myeloma (3 prior lines of treatment) was conducted      to confirm the findings of the LEGEND-2 study. Preliminary      results of the phase Ib portion showed an overall response      rate of 100% (complete response rate 86%) in patients with a      median of five prior lines of treatment (table 2).106107 The      phase II portion is fully enrolled, and phase II and III      studies have been initiated.    
      P-BCMA-101 is uniquely manufactured using the non-viral      piggyBac gene editing system, which is less costly, produces      cells with a high percentage of favorable stem cell memory      phenotype T cells, and has the ability to include a safety      switch. The binding molecule for this product is not a single      chain variable fragment but a small fully human fibronectin      domain (Centyrin) that has higher specificity and potentially      less immunogenicity. In a phase I dose escalation trial, the      overall response rate was 63% with a median progression free      survival of 9.5 months in 19 evaluable patients108 (table      2).    
      CAR T cell treatments have a unique toxicity profile where      patients can develop side effects such as cytokine release      syndrome and neurotoxicity that has been recently termed      immune effector cell associated neurotoxicity syndrome      (ICANS).116 Cytokine release      syndrome has been defined as a disorder characterized by      fever, tachypnea, headache, tachycardia, hypotension, rash,      or hypoxia caused by the release of cytokines from cells. The      American Society for Transplantation and Cellular Therapy has      developed a consensus grading system for cytokine release      syndrome, which depends on the severity and presence of      fever, hypotension, or hypoxia (table 3).116    
            American Society for Transplantation and Cellular            Therapy consensus grading for cytokine release syndrome            (CRS)116          
      ICANS has been defined as a disorder involving the central      nervous system following any immunotherapy that results in      the activation or engagement of endogenous or infused T cells      or other immune effector cells. Symptoms or signs can be      progressive and could include aphasia, altered level of      consciousness, impairment of cognitive skills, motor      weakness, seizures, and cerebral edema.116 It includes four grades that are      determined by the ICE score (immune effector cell associated      encephalopathy score, which provides objectivity to grading      encephalopathy), level of consciousness, seizure, motor      findings, and elevated intracranial pressure or cerebral      edema (table 4).116 Management of ICANS and cytokine      release syndrome is based on grading and involves supportive      care, steroids, and interleukin blocking agents.117118      Interleukin 6 blocking agents (tocilizumab and siltuximab)      with or without steroids are the mainstay of management for      cytokine release syndrome, whereas steroids are the mainstay      for the management of neurotoxicity. Another potential agent      for managing these symptoms includes the interleukin 1      blocking agent anakinra.119    
            American Society for Transplantation and Cellular            Therapy consensus grading for immune effector cell            associated neurotoxicity syndrome (ICANS) in            adults116          
      All the clinical trials on BCMA CAR T cell treatments had a      high incidence of cytokine release syndrome (>80%) except      for P-BCMA-101, which seemed to have a substantially lower      incidence (10%). Despite this, severe cytokine release      syndrome (that is, grade 3) is seen in less than 10% of      patients. Neurotoxicity was reported in less than 20% of      patients with severe neurotoxicity (grade 3) in less than 7%      of patients. Another common side effect is cytopenia, which      has also been thought to be secondary to the lymphodepleting      chemotherapy, ongoing CAR T cell activity, and disruption of      hematopoiesis showing severe hypocellularity in the bone      marrow, but most patients recover with time.120121    
      Early recognition of cytokine release syndrome and ICANS and      prompt intervention after CAR T cell treatment is vital to      prevent serious consequences, although the optimal timing for      intervention and benefit of prophylactic treatment is yet      unknown.122 The CAR T cell      therapy associated toxicity (CARTOX) working group has      developed a management approach for these syndromes, based on      multidisciplinary grades.123 In      cytokine release syndrome, patients with grade 1 are usually      managed with supportive care, those with grade 2 are managed      with the anti-interleukin 6 receptor tocilizumab with or      without steroids in addition to supportive care, and those      with grade 3-4 are managed in the intensive care unit with      aggressive supportive care, vasopressors, oxygen,      tocilizumab, and steroids. Patients with grade 1 and 2 ICANS      are managed supportively but an electroencephalogram is done      to rule out electrical seizures and imaging of the brain to      rule out edema. Patients with grade 3 and 4 ICANS need      steroids and more aggressive supportive care.120    
    Bispecific monoclonal antibodies direct a hosts immune system    (more specifically cytotoxic T cells) against cancer cells by    binding CD3 on T cells with a target protein on cancer cells    (fig 6).124 A    type of bispecific antibody is the bispecific T cell engager    (BiTE), which differs from other bispecific antibodies by    containing two different single chain variable fragments    connected by a linker. BiTEs often have a short half life,    requiring continuous infusion to maintain efficacy.125 The first BiTE to receive FDA approval    for treatment in relapsed and refractory acute lymphoblastic    leukemia is Blinatumomab, a bispecific antibody that engages T    cells to CD19 positive cells.126    Because BiTEs engage and activate the patients own immune    cells, they have a toxicity profile similar to CAR T cells    including cytokine release syndrome and ICANS.116  
            Structure of a bispecific antibody. BiTEs=bispecific T            cell engagers          
      AMG 420 (previously named BI 836909) is a novel BiTE      targeting BCMA on myeloma cells and CD3 on T cells, which      has induced multiple myeloma cell lysis in preclinical      models.127 In the      first-in-human phase I study of AMG 420 in patients with at      least two lines of treatment, AMG 420 was given as a      continuous infusion with a pump for four week infusions, six      week cycles, and a maximum of 10 cycles. The maximum      tolerated dose was 400 g/day; seven (70%) of 10 patients      responded to this dose. Serious adverse events were seen in      48% of patients, which were most commonly infections; and two      patients had reversible grade 3 polyneuropathies. Cytokine      release syndrome developed in 38% of patients, with no      toxicity in the central nervous system.109 A phase Ib trial with AMG 420 is      currently ongoing and although this drug looks promising, the      continuous intravenous infusions present logistical      challenges for patients and healthcare systems (table 2). AMG 701 is a modified      version of AMG 420 (by addition of an Fc domain) with an      extended half life that is suitable for dosing once a week      and is being investigated in a phase I study.128    
      Another BCMA bispecific antibody, CC-93269, is being studied      in an ongoing phase I clinical trial. This humanized 2+1,      immunoglobulin G 1 based, T cell engager binds to BCMA      bivalently on myeloma cells and CD3 monovalently on T cells.      The bivalent binding could lead to improved potency, tumor      targeting, and retention.129 All      doses (range 0.15-10 mg) were given intravenously over two      hours weekly for the first three cycles, every two weeks for      the next three cycles, and then monthly. The most common      treatment emergent adverse events of grade 3 or higher      included neutropenia, anemia, and infections. Cytokine      release syndrome was seen in 77% of patients, with all events      developing after the first dose and less common with      subsequent doses. The incidence increased with higher doses,      and only one patient had cytokine release syndrome of grade 3      or higher leading to their death. In 30 patients treated, the      overall response rate was 43.3% and dose dependent. The      overall response rate was 88.9% in nine patients in the      highest dose cohort.110    
      Teclistamab (JNJ-64007957) is a humanized, immunoglobulin G-4      based, bispecific DuoBody antibody that binds to BCMA and CD3      that is being studied in a phase I clinical trial. In the      dose escalation part, 78 patients received doses ranging from      0.3 g/kg to 720 g/kg. The drug is given intravenously every      week, with one to three step-up doses given within one week      before the full dose. The overall response rate was dose      dependent with no responses at doses 0.3-19.2 g/kg, 30% at      38.4-180 g/kg, and 67% at 270 g/kg. Cytokine release      syndrome was seen in 56% of patients overall and 65% patients      at doses over 38.4 g/kg. The most common adverse events at      grade 3 or higher that were related to treatment were      cytopenias and infections (table      2).111    
    Antibody drug conjugates are complex molecules composed of an    antibody that targets cancer cells and are linked to a    biologically active cytotoxic drug (known as the payload;    fig 7).125    Belantamab mafodotin (GSK2857916) is a novel humanized and    afucosylated (to improve antibody dependent cell mediated    cytotoxicity) antibody drug conjugate that targets BCMA. It    consists of an anti-BCMA monoclonal antibody conjugated to    monomethyl auristatin F, a potent microtubule inhibitor.130 This antibody drug conjugate was shown    to have selective myeloma cell killing in vitro and in vivo    thus setting the stage for clinical trials.130131  
            Structure of an antibody drug conjugate          
    This antibody was studied in a two part phase I study. The drug    was well tolerated with no dose limiting toxicities, although    corneal events (such as blurry vision, dry eyes, photophobia)    were seen in about 58% of patients; these events are a known    toxicity of monomethyl auristatin F.132    In the dose expansion phase, 35 patients were treated, and the    overall response rate was 60% with a median progression free    survival 12 months.133 In a phase II,    two arm study, the antibody was used in patients with relapsed    and refractory multiple myeloma who had failed at least three    lines of treatment. The overall response rate was 31% at the    2.5 mg/kg dose and 34% at the 3.3 mg/kg dose, which was    significantly lower than the phase I study. The corneal changes    or keratopathy were seen in 70% and 75% of patients,    respectively. Owing to the similar response rates with the 2.5    mg/kg and 3.3 mg/kg doses and a more favorable side effect    profile with the lower dose, 2.5 mg/kg will be the dose used    for future studies.112 Based on these    data, belantamab is the first anti-BCMA treatment to be FDA    approved for relapsed and refractory multiple myeloma patients    who have received four prior treatments including an anti-CD38    monoclonal antibody, a proteasome inhibitor, and an    immunomodulatory agent.  
    Preliminary results for another study with 18 patients treated    on the belantamab, bortezomib, and dexamethasone arm was    presented recently, with an overall response rate of 78%;    however, all 18 patients developed grade 1-3 keratopathy.113 This visual toxicity is a unique but    potentially serious side effect to this drug that needs close    monitoring with an ophthalmologist. Another antibody drug    conjugate, DFRF4539A, is an anti-FcRH5 (also known as FcRL5)    antibody conjugated to monomethyl auristatin and has shown    limited activity and high incidence of toxicity in a phase I    study; therefore, it was unsuccessful for this disease (table 2).134135  
Here is the original post:
Emerging immunotherapies in multiple myeloma - The BMJ