Advances in understanding the ways in which the immune system fails to control tumor growth or prevent autoimmunity have led to the development of powerful therapeutic strategies to treat these diseases. In contrast to conventional therapies that have a broadly suppressive effect, immunotherapies are more akin to targeted therapies because they are mechanistically driven and are typically developed with the goal of “drugging” a specific underlying pathway or phenotype. This means that their effects and toxicities are, at least in theory, more straightforward to anticipate. The development of functionalized antibodies, genetically engineered T cells, and immune checkpoint inhibitors continues to accelerate, illuminating new biology and bringing new treatment to patients. In the following sections, we provide an overview of immunotherapeutic concepts, highlight recent advances in the field of immunotherapies, and discuss controversies and future directions, particularly as these pertain to hematologic oncology or blood-related diseases. We conclude by illustrating how original research published in this journal fits into and contributes to the overall framework of advances in immunotherapy.

Antibody-based immunotherapy is among the most successful and well-validated treatment strategies in cancer. The mechanism of action is either direct cell killing (for example, induction of apoptosis), mimicry of basic biologic functions (recruitment of Fc-bearing effector cells that act by antibody-dependent cellular cytotoxicity [ADCC]; antibody-dependent cell phagocytosis [ADCP]; or activation of complement-dependent cytotoxicity [CDC]), or T-cell immunomodulation through blockade or activation of inhibitory or activating immune receptors. In hematologic malignancies, monoclonal antibody (mAb) targets are differentiation antigens that are expressed at distinct maturation steps of a given lineage (lineage-specific antigens [LSA]), or immunoreceptors,1  as shown in Table 1.

Table 1.

FDA-approved therapeutic mAbs and conjugates for cancer therapy

TargetmAbMOAIndicationReference
CCR4 Mogamulizumab ADCC Mycosis fungoides, Sézary syndrome (approved in 2018) 2  
CD19 Tafasitamab ADCC/ADCP DLBCL (approved 2020) 3  
CD20 Rituximab CDC/ADCC/PCD B-NHL, DLBCL, CLL, FL (approved in 1997, 2006, 2010, 2011) 4–7  
CD20 Ofatumumab CDC/ADCC/PCD CLL (approved in 2009) 8  
CD20 Obinutuzumab CDC/ADCC/PCD CLL, FL (approved in 2013, 2016) 9, 10  
CD38 Daratumumab ADCC / CDC / ADCP
Blockade of CD38 
MM (approved in 2016) 11  
CD38 Isatuximab CDC/ADCP/ADCC MM (approved in 2020) 12  
CD52 Alemtuzumab ADCC/CDC/ADCP B-CLL (approved in 2001) 13, 14  
SLAMF7 Elotuzumab ADCC MM (approved in 2015) 15  
BCMA Belantamab mafodotin-blmf ADC MM (approved in 2020) 16  
CD20 Ibritumomab tiuxetan ADC B-NHL, FL (approved in 2002) 17  
CD22 Inotuzumab ozogamicin ADC ALL (approved in 2017) 18  
CD22 Moxetumomab pasudotox ADC HCL (approved in 2018) 19  
CD30 Brentuximab vedotin ADC HL, ALCL (approved in 2011, 2018) 20, 21  
CD33 Gemtuzumab ozogamicin ADC AML (approved in 2017, 2020) 22, 23  
CD79b Polatuzumab vedotin ADC DLBCL (approved in 2019) 24  
TargetmAbMOAIndicationReference
CCR4 Mogamulizumab ADCC Mycosis fungoides, Sézary syndrome (approved in 2018) 2  
CD19 Tafasitamab ADCC/ADCP DLBCL (approved 2020) 3  
CD20 Rituximab CDC/ADCC/PCD B-NHL, DLBCL, CLL, FL (approved in 1997, 2006, 2010, 2011) 4–7  
CD20 Ofatumumab CDC/ADCC/PCD CLL (approved in 2009) 8  
CD20 Obinutuzumab CDC/ADCC/PCD CLL, FL (approved in 2013, 2016) 9, 10  
CD38 Daratumumab ADCC / CDC / ADCP
Blockade of CD38 
MM (approved in 2016) 11  
CD38 Isatuximab CDC/ADCP/ADCC MM (approved in 2020) 12  
CD52 Alemtuzumab ADCC/CDC/ADCP B-CLL (approved in 2001) 13, 14  
SLAMF7 Elotuzumab ADCC MM (approved in 2015) 15  
BCMA Belantamab mafodotin-blmf ADC MM (approved in 2020) 16  
CD20 Ibritumomab tiuxetan ADC B-NHL, FL (approved in 2002) 17  
CD22 Inotuzumab ozogamicin ADC ALL (approved in 2017) 18  
CD22 Moxetumomab pasudotox ADC HCL (approved in 2018) 19  
CD30 Brentuximab vedotin ADC HL, ALCL (approved in 2011, 2018) 20, 21  
CD33 Gemtuzumab ozogamicin ADC AML (approved in 2017, 2020) 22, 23  
CD79b Polatuzumab vedotin ADC DLBCL (approved in 2019) 24  

ADC, antibody-drug-conjugate; ALCL, anaplastic large cell lymphoma; B-NHL, B-cell non-Hodgkin lymphoma; HCL, hairy cell leukemia; MOA, mode of action; PCD, programmed cell death.

A key concept that may be obvious to the practicing hematologist/oncologist but is nevertheless worthy of attention is that mAbs generally exhibit limited efficacy as single agents. For example, the response rate of newly diagnosed diffuse large B-cell lymphoma (DLBCL) to rituximab alone is up to 37%,4  to the standard chemotherapy backbone cyclophosphamide, doxorubicin hydrochloride (hydroxydaunorubicin), vincristine sulfate (Oncovin), and prednisone (CHOP) is 69%, and to rituximab combined with CHOP is 83%.5  In chronic lymphocytic leukemia (CLL), rituximab alone, fludarabine with cyclophosphamide, and fludarabine with cyclophosphamide with rituximab lead to complete response (CR) rates of 4 to 19, 22, and 44%, respectively, and a median progression-free survival of 19-43, 32.9, and 56.8 months.25-27 Similarly, daratumumab monotherapy in relapsed/refractory multiple myeloma (R/R MM) confers an overall response rate (ORR) of 29.2%, whereas daratumumab combined with bortezomib and dexamethasone is 82.9%.28,29  These observations are particularly noteworthy in the context of the mechanism of action of these mAbs, where one would a priori expect that the combination with immunosuppressive chemotherapy should be less than additive. This paradox remains, in our view, relatively unexplained with the exception of some literature on immunogenic cell death (ICD) and depletion of immunosuppressive cells by certain chemotherapeutic agents.30-32 Interestingly, the addition of chemotherapy can also improve the antitumor response of immune checkpoint blockade,33  despite the detrimental effect of chemotherapy on the immune system (lymphopenia). A possible explanation is the induction of an adaptive immune response by ICD of tumor cells, which enhances priming and activation of cytotoxic T cells (Figure 1).

Figure 1.

Different strategies of immunotherapy in hematology. (A) Each type of immunotherapy differently affects the immune system. ADC, OV, and T cells engineered with a tumor antigen-specific TCR or CAR directly mediate an antitumor response, whereas cytokines, bispecific antibodies, immune checkpoint blockade, and therapeutic cancer vaccines stimulate endogenous immune pathways and thus indirectly induce a therapeutic effect. More specifically, immune stimulatory cytokines, such as IL-2 and interferon-α, are used to enhance the proliferation, cytotoxicity, persistence, and survival of T cells. Bispecific antibodies function as essential link between tumor cells and T cells mediating T-cell activation and tumor cell lysis. Another strategy to enhance tumor cell recognition is the ex vivo engineering of patient T cells with a CAR. Immune evasion is a common feature of tumor cells. Antibodies targeting immune checkpoints can prevent exhaustion of cytotoxic T cells and thus improve antitumor immunity. OV specifically infects tumor cells, which result in ICD. The binding of mAbs to tumor cells leads to activation of the innate immune system and tumor cells destruction. An advancement is ADCs, which specifically bind to tumor cells and induce cell death after internalization because of their cytotoxic conjugate. Therapeutic cell-based, peptide-based, or gene-based cancer vaccines induce tumor-antigen presentation by APC to boost a specific and long-lasting antitumor immune response. Antibodies or small molecules are used to neutralize immunosuppressive, tumor-derived soluble factors, such as TGF-β, IL-10, or VEGF, and thus ameliorate antitumor immunity. (B) Articles published in Blood Advances on immunology and immunotherapy in the period from November 2016 to April 2020 were classified as indicated. Most of these articles focus on cell-based immunotherapies, ICIs, and mAbs, which is in accordance with their major clinical relevance in hematology. GM-CSF, granulocyte-macrophage colony-stimulating factor; TGF-β, transforming growth factor-β; VEGF, vascular endothelial growth factor. Professional illustration by Somersault18:24.

Figure 1.

Different strategies of immunotherapy in hematology. (A) Each type of immunotherapy differently affects the immune system. ADC, OV, and T cells engineered with a tumor antigen-specific TCR or CAR directly mediate an antitumor response, whereas cytokines, bispecific antibodies, immune checkpoint blockade, and therapeutic cancer vaccines stimulate endogenous immune pathways and thus indirectly induce a therapeutic effect. More specifically, immune stimulatory cytokines, such as IL-2 and interferon-α, are used to enhance the proliferation, cytotoxicity, persistence, and survival of T cells. Bispecific antibodies function as essential link between tumor cells and T cells mediating T-cell activation and tumor cell lysis. Another strategy to enhance tumor cell recognition is the ex vivo engineering of patient T cells with a CAR. Immune evasion is a common feature of tumor cells. Antibodies targeting immune checkpoints can prevent exhaustion of cytotoxic T cells and thus improve antitumor immunity. OV specifically infects tumor cells, which result in ICD. The binding of mAbs to tumor cells leads to activation of the innate immune system and tumor cells destruction. An advancement is ADCs, which specifically bind to tumor cells and induce cell death after internalization because of their cytotoxic conjugate. Therapeutic cell-based, peptide-based, or gene-based cancer vaccines induce tumor-antigen presentation by APC to boost a specific and long-lasting antitumor immune response. Antibodies or small molecules are used to neutralize immunosuppressive, tumor-derived soluble factors, such as TGF-β, IL-10, or VEGF, and thus ameliorate antitumor immunity. (B) Articles published in Blood Advances on immunology and immunotherapy in the period from November 2016 to April 2020 were classified as indicated. Most of these articles focus on cell-based immunotherapies, ICIs, and mAbs, which is in accordance with their major clinical relevance in hematology. GM-CSF, granulocyte-macrophage colony-stimulating factor; TGF-β, transforming growth factor-β; VEGF, vascular endothelial growth factor. Professional illustration by Somersault18:24.

Close modal

Subsequently, antibody-drug conjugates (ADCs) were developed to capitalize on the property of some lineage-associated proteins to internalize upon cross-linkage, thereby delivering chemotherapy, radiation, or biologic toxins into the target cell and causing relatively specific cell death. We do not consider these drugs a form of immunotherapy because the effector mechanism is that of the toxin conjugate. However, ADCs generally have more impressive single-agent activity than naked mAbs (Table 2).

Table 2.

Therapeutic efficacy of naked mAb compared with ADC

TargetNaked mAbADC
BCMA No data available Belantamab 
 Cytotoxic payload  Monomethylauristatin F (MMAF) 
 Indication/status  R/R MM (FDA approved) 
 Median PFS (mo)  2.9 
 1-y PFS  n.d. 
 ORR, %  31 
 Reference
Clinical trial 
 16 
NCT03525678 
CD19 Tafasitamab Loncastuximab 
 Cytotoxic payload  Pyrrolobenzodiazipine (PBD) 
 Indication/status R/R B-NHL (FDA approved) R/R B-NHL (FDA approved) 
 Median PFS (mo) 2.7 (DLBCL)
8.8 (FL) 
5.5 
 1-y PFS, % 39 n.d. 
 ORR, % 29 59.4 (CR 24.1%) 
 Reference
Clinical trial 
34 
NCT01685008 
35 
NTC02669017 
CD20 Rituximab Ibritumomab 
 Cytotoxic payload  90Y 
 Indication/status R/R B-NHL (FDA approved) R/R B-NHL (FDA approved) 
 Median PFS (mo) 10.1 11.2 
 1-y PFS n.d. n.d 
 ORR, % 56 80 
 Reference 17  17  
CD22 Epratuzumab Inotuzumab 
 Cytotoxic payload  Calicheamicin 
 Indication/status R/R ALL (phase 2 study) R/R ALL (FDA approved) 
 Median OS, mo 7.7 
 1-y PFS n.d. n.d. 
 ORR, % 50 80.7 CR 
 Reference
Clinical trial 
36 
NTC01219816 
18 
NCT01564784 
CD30 SGN-30 Brentuximab (FDA approved) 
 Cytotoxic payload  Monomethylauristatin E (MMAE) 
 Indication/status R/R HL (phase 2 study) R/R HL 
 ORR, % 0 (28.9% stable disease) 86 
 Median PFS n.d. n.d. 
 2-y PFS n.d. 82.1% 
 Reference
Clinical trial 
37  38 
NCT01712490 
CD33 Lintuzumab Gemtuzumab 
 Cytotoxic payload  Calicheamicin 
 Indication/status R/R AML (phase 3 study, failed) R/R AML (FDA approved) 
 ORR, % 36 38.8 
 Median PFS Median survival 171.5 d 1 y RFS 18.5% 
 Median OS n.d. 1 y OS 26% 
 Reference
Clinical trial 
39 
NCT00006045 
40  
CD79b No clinical data available Polatuzumab 
 Cytotoxic payload  Monomethylauristatin E (MMAE) 
 Indication/status  R/R DLBCL (FDA approved) 
 Median PFS  9.5 mo 
 1-y PFS  n.d. 
 CR, %  40 
 Reference
Clinical trial 
 24 
NCT02257567 
TargetNaked mAbADC
BCMA No data available Belantamab 
 Cytotoxic payload  Monomethylauristatin F (MMAF) 
 Indication/status  R/R MM (FDA approved) 
 Median PFS (mo)  2.9 
 1-y PFS  n.d. 
 ORR, %  31 
 Reference
Clinical trial 
 16 
NCT03525678 
CD19 Tafasitamab Loncastuximab 
 Cytotoxic payload  Pyrrolobenzodiazipine (PBD) 
 Indication/status R/R B-NHL (FDA approved) R/R B-NHL (FDA approved) 
 Median PFS (mo) 2.7 (DLBCL)
8.8 (FL) 
5.5 
 1-y PFS, % 39 n.d. 
 ORR, % 29 59.4 (CR 24.1%) 
 Reference
Clinical trial 
34 
NCT01685008 
35 
NTC02669017 
CD20 Rituximab Ibritumomab 
 Cytotoxic payload  90Y 
 Indication/status R/R B-NHL (FDA approved) R/R B-NHL (FDA approved) 
 Median PFS (mo) 10.1 11.2 
 1-y PFS n.d. n.d 
 ORR, % 56 80 
 Reference 17  17  
CD22 Epratuzumab Inotuzumab 
 Cytotoxic payload  Calicheamicin 
 Indication/status R/R ALL (phase 2 study) R/R ALL (FDA approved) 
 Median OS, mo 7.7 
 1-y PFS n.d. n.d. 
 ORR, % 50 80.7 CR 
 Reference
Clinical trial 
36 
NTC01219816 
18 
NCT01564784 
CD30 SGN-30 Brentuximab (FDA approved) 
 Cytotoxic payload  Monomethylauristatin E (MMAE) 
 Indication/status R/R HL (phase 2 study) R/R HL 
 ORR, % 0 (28.9% stable disease) 86 
 Median PFS n.d. n.d. 
 2-y PFS n.d. 82.1% 
 Reference
Clinical trial 
37  38 
NCT01712490 
CD33 Lintuzumab Gemtuzumab 
 Cytotoxic payload  Calicheamicin 
 Indication/status R/R AML (phase 3 study, failed) R/R AML (FDA approved) 
 ORR, % 36 38.8 
 Median PFS Median survival 171.5 d 1 y RFS 18.5% 
 Median OS n.d. 1 y OS 26% 
 Reference
Clinical trial 
39 
NCT00006045 
40  
CD79b No clinical data available Polatuzumab 
 Cytotoxic payload  Monomethylauristatin E (MMAE) 
 Indication/status  R/R DLBCL (FDA approved) 
 Median PFS  9.5 mo 
 1-y PFS  n.d. 
 CR, %  40 
 Reference
Clinical trial 
 24 
NCT02257567 

n.d., not determined; OS, overall survival; PFS, progression-free survival; RFS, relapse-free survival; R/R ALL, relapsed/refractory acute lymphoblastic leukemia; R/R AML, relapsed/refractory acute myeloid leukemia; R/R B-NHL, relapsed/refractory B-cell non-Hodgkin lymphoma.

Two interrelated questions arise when considering this interesting class of drugs: How specific are they really, and how can they be made even more active? The side-effect profile of gemtuzumab and inotuzumab (cytopenia and hepatotoxicity) is remarkably similar given they target distinct hematopoietic lineages, brentuximab is associated with an up to 70% incidence of peripheral neuropathy,41  and belantamab mafodotin is commonly associated with keratopathy.42  These off-target toxicities suggest that future efforts to improve response using more toxic payloads would have to be done cautiously and with a detailed understanding of the conjugation chemistry, metabolism, and pharmacokinetics of the system as a whole. Novel ADCs in early-phase clinical trials include those targeting CCR7 (CLL, NHL), CD46 (R/R MM), CD71 (R/R AML, R/R ALL), CD74 (advanced B-cell malignancies), CD123 (R/R AML, R/R ALL), and ROR1 (hematologic cancer) (www.clinicaltrials.gov, searched on 12 January 2021).

mAbs have also been studied for the treatment of hematologic autoimmune diseases, particularly ones thought to be driven by autoreactive antibodies. Depletion of B cells with rituximab is surprisingly effective in some forms of autoimmune hemolytic anemia, immune thrombocytopenia, immune-mediated thrombotic thrombocytopenic purpura, treatment of inhibitors in hemophilia, and perhaps in chronic graft-versus-host disease (GVHD).43-47 With some exceptions, this approach is not particularly mechanistically driven (perhaps more opportunistic, given the widespread availability and comfort level on the part of clinicians with the use of rituximab) because most autoantibodies are likely produced by plasma cells, and indeed, in some of the abovementioned disorders (such as GVHD), the pathogenic process remains quite opaque. Furthermore, mAbs as specific neutralizers are of great interest in immune diseases that are driven primarily by the production of a cytokine, such as anti-interleukin-5 (IL-5) (mepolizumab) in hypereosinophilic syndrome,48  anti-IL-6 (siltuximab) in Castleman disease,49  and anti-interferon-γ (emapalumab) in hemophagocytic lymphohistiocystosis (HLH).50  A different method of cytokine inhibition is cytokine receptor blockade, such as IL-6R (tocilizumab; studied in the treatment of rheumatoid arthritis,51  cytokine release syndrome [CRS],52  and COVID-1953 ) or of the high-affinity IL-2 receptor (basiliximab; studied in solid-organ transplantation54  and GVHD55 ).

Alternative multispecific antigen-targeting formats, such as bispecific T-cell engaging antibodies (BiTEs), are discussed in a subsequent section because their major mechanism of action is that of the recruited effector cell.

T-cell activation is a rigorously controlled process that is dependent on signals provided by the T-cell receptor (TCR) complex and its interaction with regulatory proteins. A dynamic interplay with inhibitory and stimulatory proteins modulates the degree of T-cell activation to allow tolerance to self-antigens (inhibitory) while initiating an adaptive immune response to foreign antigens (stimulatory). Immune checkpoints are essential inhibitory stimuli, which normally maintain immune responses within a desired physiologic range by a temporary downregulation of T-cell function.

Malignant cells often coopt this protective mechanism and evade the host immune system by expressing the ligands of immune checkpoint pathways, such as CTLA-4 and PD-1 pathways. Blockade of receptors or ligands involved in these inhibitory mechanisms can in some cases reverse the tumor-mediated downregulation of T-cell function and enhance an effective antitumor immune response at the priming (CTLA-4) or tissue effector (PD-1) phase.56,57  Immunologic tolerance is retained through clonal deletion of self-reactive clones during negative selection in the thymus (central tolerance) or through peripheral tolerance, a loosely defined process that includes cell-intrinsic (eg, induction of anergy) and cell-extrinsic (regulatory T [Treg] cells, myeloid-derived suppressor cells) mechanisms. Blockade of inhibitory immune checkpoints can activate otherwise exhausted antitumor T cells. One drawback of immune checkpoint blockade is the greater probability for the activation of autoreactive T-cell clones with low signal strength that are normally incapable of generating an effective immune.58  Therefore, patients with preexisting active autoimmune disorders have historically been excluded from clinical trials using immune checkpoint inhibitors (ICIs) because of the susceptibility to develop severe adverse effects. However, mounting evidence supports the safety and effectiveness of immune checkpoint blockade in this group of patients.59 

The first promising clinical results with checkpoint blockade therapy were in the treatment of solid tumors, especially melanoma. In this malignancy, blocking of the CTLA-4 and/or PD-1 pathway has shown superior activity with a potential to induce durable response.60,61  Today, we also know that a higher mutational burden across multiple solid tumor types correlates with a greater immunogenicity and better response to ICIs.62  Hematological malignancies, however, have a very low mutational burden, which may explain the poor responses to immune checkpoint inhibition in the treatment of these diseases.63,64  We suspect however that this is not the only answer, because response rates to ICIs in hematologic malignancies with high rates of somatic mutations, such as those with mutations in TP53, do not appear to be higher.

A notable exception is Hodgkin lymphoma (HL), where alterations in 9p24.1 JAK and MEK/ERK signaling are recurrent genetic abnormalities that lead to the overexpression of PD-1 ligands (PD-L1) on Hodgkin-Reed-Sternberg cells.65  The interaction of PD-L1 with PD-1 results in dephosphorylation of proteins involved in the TCR signaling pathway, which terminates the signaling cascade and consequently inhibits T-cell activity and proliferation. Blocking PD-1 during the effector phase restores the immune function of T cells and enhances the antitumor activity, including T-cell proliferation, cytokine production, and survival.66  In clinical studies, treatment with anti–PD-1 antibodies, nivolumab or pembrolizumab, resulted in an ORR of 87% and 65% in patients with R/R Hodgkin lymphoma (R/R HL), leading to their approval in 2016 by Food and Drug Administration (FDA).67,68 

Another consideration is that the PD-1/PD-L1 axis is far from the only immune checkpoint. Following the therapeutic success of CTLA-4 and PD-1 blockade in some solid cancers, many other T-cell costimulatory molecules are now being investigated in preclinical and clinical studies. Among these molecules, LAG-3 and TIM-3 are the most advanced candidates, but there is also growing evidence for the therapeutic relevance of TIGIT and VISTA blockade to enhance antitumor immunity in hematologic disease.69,70 

LAG-3 is mainly expressed on activated CD4 and CD8 T cells, and coexpression with PD-1 correlates with an exhausted phenotype. In follicular lymphoma (FL), intratumoral PD-1+ LAG-3+ T cells were reported to be functionally suppressed, and blockade of both PD-1 and LAG-3 enhanced the functionality of intratumoral T cells.71  Currently, several phase 1/2 clinical trials are ongoing to evaluate the therapeutic potential of LAG-3 antibodies as single or in combination with PD-1 inhibitors in relapsed or refractory B-cell malignancies (NCT03489369, NCT03005782, NCT02061761). In addition, a dual-targeting antibody specific for both PD-1 and LAG-3 is investigated in a phase 1 study in patients with unresectable or metastatic neoplasms, including DLBCL (NCT03219268). An acceptable safety profile and encouraging evidence of antitumor activity have already been reported for this dual-targeting antibody.72 

More recently, it was demonstrated that the number of PD-1/TIM-3 double-positive T-cell subsets is increased in newly diagnosed and relapsed acute myeloid leukemia (AML) compared with healthy specimens.73  Interestingly, TIM-3 expression was also significantly elevated in leukemia stem cells (LSCs) and leukemic progenitors, but not in normal hematopoietic stem cells or progenitors.74  Given that LSCs are considered to be responsible for AML relapse after standard therapies, targeting of TIM-3 represents a promising novel approach in eliminating LSCs and preventing disease relapse. Therefore, TIM-3 targeting mAbs, as single agent or combined with anti–PD-1 antibodies, are currently tested in phase 1 clinical trials for both solid tumors and hematologic malignancies (NCT03489343, NCT03311412).

Another form of tumor immune evasion is the upregulation of CD47, which is a “don’t eat me” signal whose overexpression results in inhibition of phagocytosis by macrophages. CD47 is highly expressed in solid tumors and myeloid malignancies.75-78 In preclinical models of AML and myelodysplastic syndromes (MDS), CD47 blockade led to an enhanced antitumor response.77,78  In addition, anti-CD47 antibodies stimulate ADCP, thus enhancing priming and memory response of CD8 T cells.79  The therapeutic targeting of CD47 as a macrophage immune checkpoint is being investigated in several early clinical trial studies for the treatment of AML, non-Hodgkin lymphoma (NHL), MDS, HL, MM, and multiple solid tumors. A phase 1 trial of an anti-CD47 antibody in combination with azacytidine, a hypomethylating and cytotoxic agent, led to objective responses in 64% of AML patients and 92% in MDS patients with 55% or 50% achieving CR, respectively80  (NCT03248479). Although the patient number and follow-up time are limited, this report is encouraging and proves the clinical applicability of macrophage checkpoint blockade.

Although blocking antibodies have transformed cancer therapy, the development of agonist antibodies that activate costimulatory receptors to amplify antitumor immunity has been less effective. After the disastrous outcome of a CD28 superagonist antibody, which caused massive CRS and multiorgan failure in 6 healthy individuals due to T-cell activation without TCR engagement,81  current studies focus on targeting of receptors that are upregulated following T-cell activation, such as 4-1BB, OX40, GITR, CD27, and ICOS. A combination study with rituximab and utomilumab, a 4-1BB activating mAb, in 67 patients with R/R follicular lymphoma (R/R FL) and other CD20+ NHL found a 21.2% objective response rate with 4 complete and 10 partial responses. Importantly, no patient experienced dose-limiting toxicity.82  Currently, utomilumab is being tested in combination with avelumab (anti–PD-L1 mAb) in DLBCL (NCT02951156). A phase 1 study evaluated the dosage and safety of an anti-CD27 agonist antibody (varlilumab) in patients with hematologic malignancies and solid tumors (NCT01460134). In addition to T-cell stimulation, varlilumab mediates direct lysis of CD27+ lymphoma cells. However, varlilumab treatment only led to a CR in one of 10 HL patients and no objective response in 18 NHL patients (3 patients with stable disease).83  Because of this modest single-agent effect, varlilumab is further studied in combination with rituximab (NCT03307746). In this approach the induction of CDC and ADCC by rituximab is thought to be complemented by the CD27 mAb-mediated costimulation during T-cell activation.83  There are still many open questions on dosing and scheduling of agonistic antibodies, antibody structure, and combination with ICIs to improve treatment efficiency, reduce toxicity, and prevent ADCC, exhaustion, and activation-induced cell death following overstimulation of T cells with activating antibodies.

Blockade of costimulatory molecules or activation of inhibitory signaling is also of great interest for the treatment of auto- or alloimmunity. For instance, abatacept and belatacept, synthetic CTLA-4–Ig fusion proteins, are used to treat rheumatoid arthritis and improve graft survival after organ transplantation.84  An ongoing phase 2 clinical trial (NCT01743131) is investigating the addition of abatacept for the prevention of GVHD in blood cancer patients undergoing stem cell transplant. Early data showed that 6.8% of patients treated with abatacept developed severe acute GVHD compared with 14.8% in the standard treatment cohort. Importantly, abatacept addition did not increase infection risk or increase the relapse incidence. In addition, abatacept treatment was associated with a severe acute GVHD free-survival benefit (97.7% vs 58.5%).85,86  Based on these results, the FDA named abatacept a breakthrough therapy for the prevention of GVHD in hematopoietic stem cell transplants (HSCTs) from unrelated donors.

In addition to the above approaches, immune cell activity may be augmented by increasing the number of cells (stimulating proliferation ex vivo followed by adoptive transfer; administration of homeostatic or activating cytokines) or endowing them with novel functions or antigen specificity.

Ex vivo stimulation and expansion using activating beads and cytokines have been applied to effector T cells, Treg cells, and natural killer (NK) cells,87-89 whereas effector T cells have also been stimulated ex vivo with antigen-presenting cells (APCs) bearing tumor-associated or viral antigens.90-92 For the treatment of drug-refractory virus-driven cancers as well as viral infections in patients undergoing HSCT, in vitro sensitization of virus-specific T cells with viral-infected APCs or APCs loaded with infected cell lysates or synthetic peptides represents an effective therapeutic alternative. The efficiency of autologous or allogeneic Epstein-Barr virus–, cytomegalovirus-, or human papilloma virus-specific T-cell therapies is currently investigated in clinical trials, including NCT02379520, NCT02973113, and NCT03475212. Virus-specific T cells are also a potential source of allogeneic T cells for transduction with a chimeric antigen receptor (CAR). Because of the native virus-specific TCR, Epstein-Barr virus– or cytomegalovirus-specific CAR T cells have the potential of an enhanced in vivo proliferation while lacking alloreactive potential.93,94  Another interesting approach is the generation of cytokine-activated memory-like (ML) NK cells. Following ex vivo stimulation with IL-12, IL-15, and IL-18, NK cells exhibit memory-like characteristics with enhanced antitumor activity. A first-in-human phase 1 clinical trial revealed that 5 of 9 AML patients responded to adoptively transferred ML NK cells, including 4 complete remissions.95  In a second clinical trial, patients with R/R AML received a haploidentical hematopoietic cell transplant followed by same-donor ML NK cells to avoid elimination of ML NK cells by recipient allogeneic immune response. The first patients treated showed an efficient expansion and persistence (≥2 months) of highly functional ML NK cells providing first evidence for a long-term response96  (NCT02782546).

Cytokines can also be administered directly in vivo either to stimulate antitumor or to activate Treg cells.97-99 However, cytokines are typically promiscuous in their activity. IL-2 was initially developed to stimulate effector lymphocytes but is now known to stimulate Treg cells even more potently. Interesting new developments that have, as yet, to make their way into the hematologic malignancy space include synthetic cytokine/cytokine receptor pairs whose specificity can be tailored to desired cell populations. For instance, T cells engineered to express synthetic IL-2 and IL-2R, which interact with each other but not with endogenous IL-2 or IL-2R, selectively expanded and promoted an antitumor response in preclinical melanoma models.100 

Another approach is the design of chimeric antibody-cytokine fusion proteins (immunocytokines), which comprise a tumor-specific single-chain variable fragment (scFv) conjugated to an immunostimulatory cytokine, such as IL-2, IL-15, or tumor necrosis factor-α, to improve the local accumulation and pharmacokinetics compared with the native cytokines.101,102  Clinical phase 2/3 studies are currently testing the safety and activity of immunocytokines as monotherapy or in combination with ICIs for the treatment of solid tumors (NCT03420014, NCT03567889). Additional clinical investigations combine cytokines with anticancer vaccines, ICIs, and antibody-based therapies.103,104  For example, in NHL patients, recombinant IL-21 has been tested in combination with rituximab, achieving clinical response in 8 out of 19 patients.105  Further studies are currently recruiting patients to investigate the combination of recombinant IL-15 and CD20-targeting antibody therapy for the therapy of CLL (NCT03759184) or the synergistic effect of IL-15 and ICIs in relapsed/refractory mature T-cell malignancies (NCT03905135).

BiTEs can be thought of as a modality to endow effector cells with novel specificity. To date, most approaches use an anti-CD3ε antibody fragment as T-cell engaging domain fused to a scFv targeting a tumor-associated antigen. Binding of the BiTE to both targets, the TCR complex and the tumor antigen, mediates the formation of a cytolytic synapse resembling natural immunological synapses. Thus far, blinatumomab, a CD3×CD19 BiTE, is the only BiTE with FDA approval and is used for the treatment R/R B-cell precursor ALL (pre–B-ALL). Although blinatumomab showed only sustained responses, impressive results in phase 1 and 2 studies for R/R DLBCL, benefits in response duration have been reported and are further investigated in ongoing phase 1/2 trials.106  At this time, blinatumomab is not approved for NHL because of a lack of phase 3 trials, difficult mode of administration, and availability of alternative options with similar therapeutic efficacy.107  Notably, blinatumomab is particularly active in ALL patients with the minimal residual disease where its CR rate is 80%, whereas in the setting of active disease, the CR rate is 43%.108,109  Not surprisingly, blinatumomab is now being tested in combination with ICIs. One study of blinatumomab in combination with pembrolizumab reported an acceptable toxicity and a 50% ORR in adults with heavily pretreated R/R B-ALL.110 

To improve the feasibility and/or efficacy of the BiTE platform, novel CD19×CD3 constructs have been developed with improved half-life and higher CD3 affinity. Two candidates, AFM11 and MGD011, have been tested in patients with NHL and CLL or NHL and ALL, but their clinical development was discontinued because of high levels of neurotoxicity.111,112  An alternative target in B-cell malignancies is CD20, and several CD20×CD3 BiTEs are tested in ongoing clinical trials (Table 3). In an early-phase clinical trial, treatment with single-agent mosunetuzumab, a fully humanized CD20×CD3 bispecific antibody, induced durable responses in patients with B-cell NHL, even in those who relapsed following CAR T-cell therapy.113  Treatment-associated adverse events were similar to those typically observed with CAR T-cell therapy with CRS in 28.9% and neurologic toxicity in 43.7% of patients.

Table 3.

Overview of immune cell redirecting bispecific antibodies investigated in ongoing clinical trials for hematologic malignancies

TargetMolecule/drugConditionPhase; status; NCT#
BCMA + CD3 JNJ-64007957 (Teclistamab) MM 1; recruiting; NCT03145181
1; recruiting; NCT04696809
2; recruiting; NCT04557098 
BMCA + CD3 PF-06863135 (Elranatamab) MM 1; recruiting; NCT03269136
2; recruiting; NCT04649359 
BCMA + CD3 plus
GPRC5D + CD3 
JNJ-64007957 (Teclistamab); JNJ-64407564 (Talquetamab) MM 1; recruiting; NCT04108195
1; recruiting; NCT04586426 
BCMA + CD3 TNB-383B MM 1; recruiting; NCT03933735 
BCMA + CD3 REGN5458 MM 1/2; recruiting; NCT03761108 
BCMA + CD3 REGN5459 MM 1; recruiting; NCT04083534 
CD19 + CD3 TNB-486 B-cell lymphoma, FL 1; recruiting; NCT04594642 
CD20 + CD3 REGN1979 (Odronextamab) NHL, HL, CLL 1; active, not recruiting; NCT02651662, NCT02290951
2; active, not recruiting; NCT03888105 
CD20 + CD3 RG7828 (Mosunetuzumab) NHL, CLL 1; recruiting; NCT02500407, NCT03671018, NCT03677141, NCT03677154 
CD33 + CD3 AMG330 AML 1; recruiting; NCT02520427, NCT02106091 
CD33 + CD3 AMV564 AML, MDS 1; active, not recruiting; NCT03516591, NCT03144245 
CD33 + CD3 GEM333 AML 1; active, not recruiting; NCT03516760 
CD33 + CD3 JNJ-67371244 AML, MDS 1; recruiting; NCT03915379 
CD33 + CD3 (with IL-15 crosslinker) 161533 TriKE MDS, AML, ASM 1/2; recruiting; NCT03214666 
CD38 + CD3 ISB 1342 MM 1/2; recruiting; NCT03309111 
CD123 + CD3 MGD006 (Flotetuzumab) AML, MDS 1; recruiting; NCT02152956
1; recruiting; NCT04681105 
CD123 + CD3 APVO436 AML, MDS 1; recruiting; NCT03647800 
CD123 + CD3 JNJ-63709178 AML 1; recruiting; NCT02715011 
CD123 + CD3 Xmab14045 (Vibecotamab) AML, B-cell ALL, BPDCN, CML 1; recruiting; NCT02730312 
GPRC5D + CD3 JNJ-64407564
(Talquetamab) 
Hematological malignancies 1; recruiting; NCT03399799
1; recruiting; NCT04634552 
TargetMolecule/drugConditionPhase; status; NCT#
BCMA + CD3 JNJ-64007957 (Teclistamab) MM 1; recruiting; NCT03145181
1; recruiting; NCT04696809
2; recruiting; NCT04557098 
BMCA + CD3 PF-06863135 (Elranatamab) MM 1; recruiting; NCT03269136
2; recruiting; NCT04649359 
BCMA + CD3 plus
GPRC5D + CD3 
JNJ-64007957 (Teclistamab); JNJ-64407564 (Talquetamab) MM 1; recruiting; NCT04108195
1; recruiting; NCT04586426 
BCMA + CD3 TNB-383B MM 1; recruiting; NCT03933735 
BCMA + CD3 REGN5458 MM 1/2; recruiting; NCT03761108 
BCMA + CD3 REGN5459 MM 1; recruiting; NCT04083534 
CD19 + CD3 TNB-486 B-cell lymphoma, FL 1; recruiting; NCT04594642 
CD20 + CD3 REGN1979 (Odronextamab) NHL, HL, CLL 1; active, not recruiting; NCT02651662, NCT02290951
2; active, not recruiting; NCT03888105 
CD20 + CD3 RG7828 (Mosunetuzumab) NHL, CLL 1; recruiting; NCT02500407, NCT03671018, NCT03677141, NCT03677154 
CD33 + CD3 AMG330 AML 1; recruiting; NCT02520427, NCT02106091 
CD33 + CD3 AMV564 AML, MDS 1; active, not recruiting; NCT03516591, NCT03144245 
CD33 + CD3 GEM333 AML 1; active, not recruiting; NCT03516760 
CD33 + CD3 JNJ-67371244 AML, MDS 1; recruiting; NCT03915379 
CD33 + CD3 (with IL-15 crosslinker) 161533 TriKE MDS, AML, ASM 1/2; recruiting; NCT03214666 
CD38 + CD3 ISB 1342 MM 1/2; recruiting; NCT03309111 
CD123 + CD3 MGD006 (Flotetuzumab) AML, MDS 1; recruiting; NCT02152956
1; recruiting; NCT04681105 
CD123 + CD3 APVO436 AML, MDS 1; recruiting; NCT03647800 
CD123 + CD3 JNJ-63709178 AML 1; recruiting; NCT02715011 
CD123 + CD3 Xmab14045 (Vibecotamab) AML, B-cell ALL, BPDCN, CML 1; recruiting; NCT02730312 
GPRC5D + CD3 JNJ-64407564
(Talquetamab) 
Hematological malignancies 1; recruiting; NCT03399799
1; recruiting; NCT04634552 

ASM, advanced systemic mastocytosis; BPDCN, blastic plasmacytoid dendritic cell neoplasm; CML, chronic myeloid leukemia; GPRC5D, G-protein–coupled receptor family C group 5 member D; TriKE, trispecific killer engager.

In the treatment of MM, daratumumab and belantamab have been shown to be effective; therefore, several BiTE constructs targeting CD38 or BCMA are currently evaluated in early-phase clinical trials (Table 3). An interesting construct in the treatment of HL is AFM13, a tetravalent bispecific antibody activating the innate immune system (CD30×CD16). In a phase 1 trial, 3 out of 26 patients with R/R HL achieved a partial response and 13 patients achieved a stable disease.114  The antibody was well tolerated (grade 3 AE 9%) and tested in phase 2 trials in R/R HL (NCT02321592), cutaneous lymphoma (NCT03192202), and in combination with pembrolizumab in R/R HL (NCT02665650).

In AML, several BiTEs have been developed, and phase 1 trials with CD33×CD3 or CD123×CD3 are ongoing (Table 3). First reports of flotetuzumab, a CD123×CD3 bispecific antibody-derived molecule, in patients with AML and high-risk MDS, showed a significant increase of CD8 T cells in bone marrow samples and an ORR of 43% with manageable toxicity (grade ≥3 CRS 13%)115  (NCT02152956).

Remarkably, the reasons for primary or secondary failure of BiTEs are relatively underexplored. Therefore, combining BiTE with ICIs may play a key role in further advancing BiTE therapy. Another tumor-resistance mechanism is the downregulation of targeted antigens. Loss of CD19 expression following blinatumomab treatment is reported in 8% to 30% of relapsed ALL patients.116,117  This issue could be addressed by the design of multivalent BiTE molecules, which enhance target avidity as well as trispecific antibodies that recognize >1 tumor antigen.118 

Novel functions can be conferred on effector T cells, NK cells, or macrophages through genetic means. T cells can be engineered to express a transgenic TCR specific for a tumor-associated antigen in the context of the relevant major histocompatibility (MHC) molecule or a CAR specific for a cell-surface target. Most tumor-driving mutations or overexpressed tumor-associated antigens are located intracellularly and therefore are not accessible to CAR T cells. However, the major limitations to date of TCR transgenic T cells are the requirement for MHC presentation and matching (particularly important because MHC downregulation is a common tumor immune evasion mechanism), technical difficulties with expression of costimulatory molecules, and the need to replace or delete the endogenous α/β TCR chains in order to reduce the chance of mispairing leading to novel (and potentially pathogenic) specificities.119,120 

To date, 4 CD19-specific CAR T-cell therapies, axicabtagene ciloleucel, tisagenlecleucel, brexucabtagene autoleucel, and lisocabtagene maraleucel, have been approved for the treatment of B-ALL and certain B-cell lymphoma,121-125 and 1 B-cell maturation antigen (BCMA)-directed CAR T-cell product (idecabtagene vicleucel) for MM.126  Numerous clinical trials are currently ongoing to investigate CAR T cells for the treatment of hematologic neoplasms. The most promising targets for these studies are CD19, CD20, CD22, and BCMA.127  TCR T-cell products have not yet been approved, but clinical trials are evaluating the safety and therapeutic activity of HA-1 and WT1-specific TCR T cells in AML, MDS, and ALL patients (NCT03326921, NCT02770820).

Although CAR T-cell therapy is approved by the FDA as standard of care for some forms of aggressive, relapsed, or refractory hematologic malignancies, there are several challenges and hurdles that must be overcome to enable the widespread use of CAR T-cell therapy. Key challenges that we think are reasonably well understood include CRS, antigen escape, and manufacturing logistics. Important challenges that we think remain poorly understood in the hematologic malignancies space include neurotoxicity, trafficking, interactions with the tumor microenvironment, and limited in vivo persistence.128,129  Preclinical and clinical studies are addressing this wide range of obstacles in order to broaden the applicability of CAR T-cell therapy. The risk of side effects, including CRS and neurotoxicity, correlates with tumor burden, the dose of infused CAR T cells, CAR design, and patient factors, such as age and preexisting comorbidities. The understanding of CAR-mediated pathophysiology is increasing, and predictive models for the detection and prevention of CRS or neurotoxicity are currently developed but require further validation. So far, toxicity management includes supportive care and immunosuppression with tocilizumab and sometimes corticosteroids.130  Experimental approaches to reduce toxicity and enhance the safety of CAR T-cell therapy include prevention of on-target/off-tumor effects by selecting appropriate target antigens or controlling CAR activity by suicide genes or switch-off designs. In the case of CD19-specific CAR T cell, the targeting results in healthy B cells and subsequent B-cell aplasia and hypogammaglobulinemia, which may require administration of IV immunoglobulin.131,132  Diminished proliferation, persistence, and antitumor capacity can be enhanced by engineering T cells with a less-differentiated phenotype.133  One attempt to address antigen escape and tumor heterogeneity is the combinatorial targeting of multiple antigens by BiTE-secreting CAR T cells or multispecific CAR T cells.134,135  Another well-known disadvantage of autologous CAR T-cell therapy is the costly and time-consuming manufacturing process leading to treatment delay, which is particularly problematic for patients with highly proliferative diseases. Current research uses novel gene-editing tools, such as TALEN and CRISPR-Cas9, to further refine adoptive cell therapy approaches. Such genetic tools for overexpression (viral or transposon based) or deletion (CRISPR, TALEN, or ZFN) of selected molecules allow the depletion of immune checkpoints to enhance antitumor activity or replacement the endogenous TCR with a tumor-specific TCR or CAR for the generation of allogeneic, off-the-shelf cell products.136-138 Although gene editing holds immense potential, limitations for clinical application include off-target editing events, low efficiency, and impaired expansion of ex vivo edited cells. Furthermore, most gene-editing tools have only been tested in vitro, and an open question is the safety and efficacy in clinical settings.139  Therefore, the combination of already approved drugs, such as ICIs and adoptive cell therapy, holds promise for more rapid clinical implementation. For instance, a small, single-center study at Children’s Hospital of Pennsylvania reported an improved persistence of CD19-CAR T cells because of the addition of PD-1 blockade in children with heavily pretreated B-ALL, including allo-SCT.140  Other studies testing the combination of ICIs with CAR T cells are in progress, and results are eagerly awaited. An alternative approach to enhance the efficacy and persistence of CAR T cells is the coexpression of cytokines, such as IL-12 or IL-15, or cytokine receptors that stimulate T-cell proliferation.141-143 An ongoing phase 1 trial investigates the safety and dose of CD19 CAR T cells coexpressing membrane-bound IL-15 in patients with lymphoma (NCT03579888).

In addition to therapeutic applications in oncology, bispecific antibodies and cell-based therapies have been developed for the treatment of inflammatory disorders. In 2017, the bispecific antibody emicizumab was approved for the treatment of acquired hemophilia A, a severe bleeding disorder caused by inhibitory autoantibodies to coagulation factor VIII. Emicizumab bridges activated factor IX and factor X to restore the function of missing activated factor VIII, which is needed for effective hemostasis.144  The CAR concept has also been translated to treat transplant rejection and autoimmunity. Studies reported that CAR-expressing Treg cells mediated a therapeutic benefit in mouse models of colitis, multiple sclerosis, GVHD, and islet and skin transplantation.145-147 Recently, the safety of CAR-modified Treg cells was shown in phase 1 clinical trials.148,149  Another interesting approach is the replacement of the traditional extracellular scFv with an autoantigen to target autoreactive B cells with engineered T cells, which may be a strategy to treat antibody-mediated autoimmune disease.150 

Despite the tantalizing potential of vaccines that is clearly illustrated in the world of infectious diseases,151  including the recent severe acute respiratory syndrome coronavirus 2 vaccine,152  cancer cells are not foreign microorganisms, and the development of effective cancer vaccines still represents a major challenge for the field.

To date, only 2 therapeutic cancer vaccines have been approved by the FDA, sipuleucel-T (Provenge) and Bacillus Calmette-Guerin, which are indicated for the treatment of metastatic castration-resistant prostate cancer or nonmuscle-invasive bladder cancer, and unfortunately, show marginal efficacy.153,154 

A major focus of the development of therapeutic cancer vaccines is the selection of optimal target antigens, which are aberrantly expressed self-antigens. Because high-affinity T cells that recognize self-antigens are deleted at an early stage of lymphoid cell development (central tolerance), therapeutic cancer vaccines face the challenge of activating any remaining, low-affinity T cells. New strategies are developed for the selection of more immunogenic tumor-associated self-antigens and neoantigens that harbor tumor-specific mutations to improve cancer vaccines. A pilot trial of a WT1-targeting multivalent heteroclitic peptide vaccine (galinpepimut-S) following autologous stem cell transplantation found a favorable safety profile and a robust CD4 and CD8 T-cell response in 16 high-risk MM patients, which led to an encouraging median progression-free survival of 23.6 months.155  A phase 1/2 clinical trial is currently recruiting patients to assess the combination of galinpepimut-S and pembrolizumab in patients with advanced cancers including AML (NCT03761914).

In therapeutic settings, a vaccine-stimulated immune response is challenged by a high tumor burden with established immunoregulatory pathways to dampen a natural immune response. Therefore, enhanced vaccine technologies include costimulatory components, such as adjuvants, cytokines, or other agents, that improve the efficacy of cancer vaccines. Delivery of an antigen without appropriate costimulator can result in T-cell ignorance, T-cell anergy, or even T-cell deletion.156  In addition, combinations with ICIs and other new drugs that reverse immunosuppression are showing promising results in preclinical studies.157,158  This success, however, has not yet been translated into clinical benefits. Although early-phase clinical trials show the feasibility and tolerability of cancer vaccines combined with ICIs, the immunologic effect is only marginal and does not lead to a significant improvement in overall survival when compared with ICI monotherapy.159-161 Further studies are needed to clarify whether this combination therapy has a synergistic effect and can improve patient outcome.

In contrast to therapeutic vaccines, prophylactic vaccines are more successful, and several vaccines have been approved to prevent hepatitis B virus and human papilloma virus infection, which are associated with liver and cervical cancer.162  To our knowledge, there are no data on vaccines to prevent virally driven hematological malignancies.

Vaccination strategies are also effective as allergen-specific immunotherapy. Administration of protein/peptide-based allergens in repeated and often escalating doses prevents disease progression or, in some cases, provides a curative therapy in individuals with allergic rhinitis or asthma. We are not aware of any work using tolerogenic vaccines in autoimmune hematological diseases.

Oncolytic viruses (OVs) have the potential to specifically infect tumor cells and induce ICD, which may result in a potent and long-lasting antitumor response.163  In 2015, the first OV therapy, talimogene laherparepvec, was approved by the FDA for the treatment of nonresectable metastatic melanoma. Talimogene laherparepvec is a modified herpes simplex virus-1 that was genetically modified to express granulocyte-macrophage colony-stimulating factor (GM-CSF) combining virus-mediated cytotoxicity with immune stimulation.164  Hematologic malignancies, however, still represent a therapeutic challenge for OV therapy given the immune response to viral infections. Intravascular administration of OVs poses the risk of an excessive immune activation with CRS, systemic inflammatory response syndrome, and multiorgan failure. In the other extreme, there is the potential for rapid clearance of OVs and an ineffective dosing after intravascular delivery, illustrating why intratumoral delivery is commonly used for OVs.165,166  In addition, not all viruses are suitable for treatment of hematological neoplasms. Adenovirus, which is one of the most studied OVs, is described to be unable to lyse white blood cells,167  and many humans have neutralizing antibodies against different adenovirus serotypes, which makes a systemic administration ineffective.168,169  An interesting approach is the combination of an OV with CAR T cells to overcome the heterogeneous tumor antigen expression in solid tumors. Park et al designed an OV encoding truncated CD19, which led to a specific and stable target expression and enhanced tumor cell killing following treatment with CD19-specific CAR T cells in preclinical models.170 

To date, only a few trials on OV for cancer immunotherapy have been published.171  A search from clinicaltrials.gov (14 October 2020) found 38 actively recruiting studies for OV therapy in solid tumors, but only 1 phase 1 study in hematological neoplasms that currently evaluates the combination of PD-1 blockade and OV therapy in in MM (NCT03605719).

Concluding remarks and the contributions made by Blood Advances to immunotherapy in hematology

Immunotherapy has dramatically changed the quality of life and survival of some patients. Despite rapid advances in the last decade, in our view, the field remains in an “exponential growth phase” with a ferment of basic and translational research that will likely validate the promise and help us to navigate the perils of immunotherapy (see information box below).

The holy grail of immunotherapy is as a “one-and-done” intervention, if we can discover which keys to press in order to activate the patient’s faltering endogenous immune system. However, real-world examples of this in hematology are essentially limited to long-term disease-free survival in some patients with B-cell malignancies who received a single infusion of CAR T cells directed against CD19,172  and the biological and clinical correlates of even this small group of patients remain frustratingly opaque. Short of this lofty goal, immunotherapy can and does provide an additional weapon in the therapeutic arsenal for patients whose disease has not responded to more “traditional” measures, because the toxicity and vulnerability profiles are often nonoverlapping with those of chemotherapy, radiation, surgery, transplantation, or immunosuppressants. For example, antitumor mAbs are exquisitely specific and typically well tolerated (although exhibit limited single-agent efficacy), leading to fruitful combinations with chemotherapy. Validation and experience with mAb technology paved the way to bispecific T-cell (or other effector cell) engaging antibodies, which exhibit somewhat higher single-agent activity, and to the use of mAbs to block or activate immune circuits such as the PD/PD-L1 axis. High response rates to CAR T cells in hematologic malignancies ignited efforts to genetically engineer other effector or regulatory cells in increasingly sophisticated attempts to “hack” pathobiology. The high cost of novel immunotherapies (most especially CAR T cells) is often tallied against them; however, if these therapies are curative, the high initial outlay may well prove cost-efficient in the long term because the patient would be able to avoid chronic or sequential therapy with other novel (and noncurative) agents. Another interesting observation with increasing scientific interest is the role of the gut microbiome and the response to immunotherapy and development of autoimmune disease.173,174  A study of PD-1 blockade found that a high gut microbiome diversity enhanced antitumor immune response by increased antigen presentation and improved effector T-cell functionality in the tumor microenvironment.175  However, further research is needed to identify immunostimulatory and immunosuppressive bacteria species in the gut and define their effect on responsiveness to immunotherapy. Further knowledge on the connection of the microbiome and the immune system represents a unique opportunity to activate or suppress immunity for therapeutic purposes.

The perils of immunotherapy extend beyond their well-described and increasingly well-understood side effects.176-179 We think it is crucial to understand mechanisms of response as well as failure in order to avoid expending human and material capital on novel agents that are poorly conceived or lack a therapeutic rationale. For example, PD-L1 expression is a frequent immune evasion mechanism, and blocking antibodies can enhance antitumor immunity in some solid cancers. However, other than in HL, this axis has not yet been fruitfully targeted in hematologic malignancies, and we think a sober explanation of this observation is warranted. Old dogmas should be revisited: early enthusiasm for therapeutic cytokine administration (eg, IL-2 in patients with melanoma) was dampened by negative clinical outcomes in hematology, but we now understand that IL-2 can also stimulate counterregulatory and immunosuppressive cells, and novel approaches to delivering this and other cytokines have been devised.100,142,180,181  Cancer vaccines illustrate a different peril. Vaccines are highly effective at preventing certain infectious diseases but rarely effective in treating active infections; this issue may well be more acute in the immunosuppressive cancer environment, and here, the peril of immunotherapy may be in a “type II error” wherein a potentially active treatment modality is not given the opportunity to show its full potential because it is not tested in the right setting. Indeed, AML vaccines have shown promising efficacy in patients in remission.182 

Future directions in immunotherapy for hematological diseases

  • To understand the correlates of response and failure in order to predict which patients should receive what treatment, and to define suitable next-line therapies

  • To understand the mechanisms of off-target toxicity and whether these can be dissociated from efficacy

  • To identify cancer-specific targets to reduce on-target toxicity

  • Logistics and cost considerations

  • Rational combinations

Immunotherapy is a fast-growing field of research reflected by the large number of ongoing preclinical and clinical studies evaluating novel treatment strategies and therapeutic combinations with the aim to enhance effectivity, safety, and applicability. New findings on these topics and further research to understand the complexity of the immune system have been reported in Blood Advances in recent years, thereby contributing to the success story of immunotherapy. Most of these publications (41%) focused on the enhancement of immune cells (with two-thirds of the articles in the context of cancer therapy and one-third of the articles in the context of hyperimmunity and prevention thereof, including GVHD, HSCT, hemophilia). Sixteen percent of the articles reported novel findings on antibody-mediated immune response (three-fourths in cancer and one-fourth in autoimmunity), and 7% of the articles reported immune checkpoint inhibition. These and other developments will further clarify the role of immunotherapy in cancer and autoimmune disease within the next decade.

This work was supported in part by a Leukemia and Lymphoma Society Specialized Center of Research Grant (S.G.) and by the Mark Foundation (S.L.).

Contribution: S.L. and S.G. wrote and revised the manuscript; both authors approved the final manuscript.

Conflict-of-interest disclosure: S.G. holds multiple patents for CAR T cell-related research. S.L. declares no competing financial interests.

Correspondence: Saar Gill, University of Pennsylvania, Smilow Center for Translational Research, Room 8-101, 3400 Civic Center Boulevard, Philadelphia, PA 19104; e-mail: saargill@pennmedicine.upenn.edu.

1.
Scott
AM
,
Allison
JP
,
Wolchok
JD.
Monoclonal antibodies in cancer therapy
.
Cancer Immun.
2012
;
12
:
14
.
2.
Kasamon
YL
,
Chen
H
,
de Claro
RA
, et al
.
FDA approval summary: mogamulizumab-kpkc for mycosis fungoides and Sézary syndrome
.
Clin Cancer Res.
2019
;
25
(
24
):
7275
-
7280
.
3.
Salles
G
,
Duell
J
,
González Barca
E
, et al
.
Tafasitamab plus lenalidomide in relapsed or refractory diffuse large B-cell lymphoma (L-MIND): a multicentre, prospective, single-arm, phase 2 study
.
Lancet Oncol.
2020
;
21
(
7
):
978
-
988
.
4.
Coiffier
B
,
Haioun
C
,
Ketterer
N
, et al
.
Rituximab (anti-CD20 monoclonal antibody) for the treatment of patients with relapsing or refractory aggressive lymphoma: a multicenter phase II study
.
Blood.
1998
;
92
(
6
):
1927
-
1932
.
5.
Coiffier
B
,
Lepage
E
,
Briere
J
, et al
.
CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma
.
N Engl J Med.
2002
;
346
(
4
):
235
-
242
.
6.
Casak
SJ
,
Lemery
SJ
,
Shen
YL
, et al
.
U.S. Food and Drug Administration approval: rituximab in combination with fludarabine and cyclophosphamide for the treatment of patients with chronic lymphocytic leukemia
.
Oncologist.
2011
;
16
(
1
):
97
-
104
.
7.
Salles
G
,
Seymour
JF
,
Offner
F
, et al
.
Rituximab maintenance for 2 years in patients with high tumour burden follicular lymphoma responding to rituximab plus chemotherapy (PRIMA): a phase 3, randomised controlled trial
.
Lancet.
2011
;
377
(
9759
):
42
-
51
.
8.
Lemery
SJ
,
Zhang
J
,
Rothmann
MD
, et al
.
U.S. Food and Drug Administration approval: ofatumumab for the treatment of patients with chronic lymphocytic leukemia refractory to fludarabine and alemtuzumab
.
Clin Cancer Res.
2010
;
16
(
17
):
4331
-
4338
.
9.
Lee
HZ
,
Miller
BW
,
Kwitkowski
VE
, et al
.
U.S. Food and Drug Administration approval: obinutuzumab in combination with chlorambucil for the treatment of previously untreated chronic lymphocytic leukemia
.
Clin Cancer Res.
2014
;
20
(
15
):
3902
-
3907
.
10.
Sehn
LH
,
Chua
N
,
Mayer
J
, et al
.
Obinutuzumab plus bendamustine versus bendamustine monotherapy in patients with rituximab-refractory indolent non-Hodgkin lymphoma (GADOLIN): a randomised, controlled, open-label, multicentre, phase 3 trial
.
Lancet Oncol.
2016
;
17
(
8
):
1081
-
1093
.
11.
Bhatnagar
V
,
Gormley
NJ
,
Luo
L
, et al
.
FDA approval summary: daratumumab for treatment of multiple myeloma after one prior therapy
.
Oncologist.
2017
;
22
(
11
):
1347
-
1353
.
12.
Attal
M
,
Richardson
PG
,
Rajkumar
SV
, et al;
ICARIA-MM study group
.
Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study
.
Lancet.
2019
;
394
(
10214
):
2096
-
2107
.
13.
Hillmen
P
,
Skotnicki
AB
,
Robak
T
, et al
.
Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia
.
J Clin Oncol.
2007
;
25
(
35
):
5616
-
5623
.
14.
Keating
MJ
,
Flinn
I
,
Jain
V
, et al
.
Therapeutic role of alemtuzumab (Campath-1H) in patients who have failed fludarabine: results of a large international study
.
Blood.
2002
;
99
(
10
):
3554
-
3561
.
15.
Lonial
S
,
Dimopoulos
M
,
Palumbo
A
, et al;
ELOQUENT-2 Investigators
.
Elotuzumab therapy for relapsed or refractory multiple myeloma
.
N Engl J Med.
2015
;
373
(
7
):
621
-
631
.
16.
Lonial
S
,
Lee
HC
,
Badros
A
, et al
.
Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): a two-arm, randomised, open-label, phase 2 study
.
Lancet Oncol.
2020
;
21
(
2
):
207
-
221
.
17.
Witzig
TE
,
Gordon
LI
,
Cabanillas
F
, et al
.
Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin’s lymphoma
.
J Clin Oncol.
2002
;
20
(
10
):
2453
-
2463
.
18.
Kantarjian
HM
,
DeAngelo
DJ
,
Stelljes
M
, et al
.
Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia
.
N Engl J Med.
2016
;
375
(
8
):
740
-
753
.
19.
Kreitman
RJ
,
Dearden
C
,
Zinzani
PL
, et al
.
Moxetumomab pasudotox in relapsed/refractory hairy cell leukemia
.
Leukemia.
2018
;
32
(
8
):
1768
-
1777
.
20.
Horwitz
S
,
O’Connor
OA
,
Pro
B
, et al;
ECHELON-2 Study Group
.
Brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma (ECHELON-2): a global, double-blind, randomised, phase 3 trial
.
Lancet.
2019
;
393
(
10168
):
229
-
240
.
21.
Straus
DJ
,
Długosz-Danecka
M
,
Alekseev
S
, et al
.
Brentuximab vedotin with chemotherapy for stage III/IV classical Hodgkin lymphoma: 3-year update of the ECHELON-1 study
.
Blood.
2020
;
135
(
10
):
735
-
742
.
22.
Castaigne
S
,
Pautas
C
,
Terré
C
, et al;
Acute Leukemia French Association
.
Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study
.
Lancet.
2012
;
379
(
9825
):
1508
-
1516
.
23.
FDA approves gemtuzumab ozogamicin for CD33-positive AML in pediatric patients
, https://bit.ly/37xYqh8 (
2020
).
24.
Sehn
LH
,
Herrera
AF
,
Flowers
CR
, et al
.
Polatuzumab vedotin in relapsed or refractory diffuse large B-cell lymphoma
.
J Clin Oncol.
2020
;
38
(
2
):
155
-
165
.
25.
Fischer
K
,
Bahlo
J
,
Fink
AM
, et al
.
Long-term remissions after FCR chemoimmunotherapy in previously untreated patients with CLL: updated results of the CLL8 trial
.
Blood.
2016
;
127
(
2
):
208
-
215
.
26.
Hainsworth
JD
,
Litchy
S
,
Barton
JH
, et al;
Minnie Pearl Cancer Research Network
.
Single-agent rituximab as first-line and maintenance treatment for patients with chronic lymphocytic leukemia or small lymphocytic lymphoma: a phase II trial of the Minnie Pearl Cancer Research Network
.
J Clin Oncol.
2003
;
21
(
9
):
1746
-
1751
.
27.
Hallek
M
,
Fischer
K
,
Fingerle-Rowson
G
, et al;
German Chronic Lymphocytic Leukaemia Study Group
.
Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial
.
Lancet.
2010
;
376
(
9747
):
1164
-
1174
.
28.
Lonial
S
,
Weiss
BM
,
Usmani
SZ
, et al
.
Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial
.
Lancet.
2016
;
387
(
10027
):
1551
-
1560
.
29.
Palumbo
A
,
Chanan-Khan
A
,
Weisel
K
, et al;
CASTOR Investigators
.
Daratumumab, bortezomib, and dexamethasone for multiple myeloma
.
N Engl J Med.
2016
;
375
(
8
):
754
-
766
.
30.
Alizadeh
D
,
Trad
M
,
Hanke
NT
, et al
.
Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer
.
Cancer Res.
2014
;
74
(
1
):
104
-
118
.
31.
Galluzzi
L
,
Buqué
A
,
Kepp
O
,
Zitvogel
L
,
Kroemer
G.
Immunogenic cell death in cancer and infectious disease
.
Nat Rev Immunol.
2017
;
17
(
2
):
97
-
111
.
32.
Ghiringhelli
F
,
Menard
C
,
Puig
PE
, et al
.
Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients
.
Cancer Immunol Immunother.
2007
;
56
(
5
):
641
-
648
.
33.
Paz-Ares
L
,
Ciuleanu
TE
,
Cobo
M
, et al
.
First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): an international, randomised, open-label, phase 3 trial
.
Lancet Oncol.
2021
;
22
(
2
):
198
-
211
.
34.
Jurczak
W
,
Zinzani
PL
,
Gaidano
G
, et al
.
Phase IIa study of the CD19 antibody MOR208 in patients with relapsed or refractory B-cell non-Hodgkin’s lymphoma
.
Ann Oncol.
2018
;
29
(
5
):
1266
-
1272
.
35.
Kahl
BS
,
Hamadani
M
,
Radford
J
, et al
.
A phase I study of ADCT-402 (loncastuximab tesirine), a novel pyrrolobenzodiazepine-based antibody-drug conjugate, in relapsed/refractory B-cell non-Hodgkin lymphoma
.
Clin Cancer Res.
2019
;
25
(
23
):
6986
-
6994
.
36.
Chevallier
P
,
Chantepie
S
,
Huguet
F
, et al
.
Hyper-CVAD + epratuzumab as a salvage regimen for younger patients with relapsed/refractory CD22-positive precursor B-cell acute lymphocytic leukemia
.
Haematologica.
2017
;
102
(
5
):
e184
-
e186
.
37.
Forero-Torres
A
,
Leonard
JP
,
Younes
A
, et al
.
A phase II study of SGN-30 (anti-CD30 mAb) in Hodgkin lymphoma or systemic anaplastic large cell lymphoma
.
Br J Haematol.
2009
;
146
(
2
):
171
-
179
.
38.
Connors
JM
,
Jurczak
W
,
Straus
DJ
, et al;
ECHELON-1 Study Group
.
Brentuximab vedotin with chemotherapy for stage III or IV Hodgkin’s lymphoma
.
N Engl J Med.
2018
;
378
(
4
):
331
-
344
.
39.
Feldman
EJ
,
Brandwein
J
,
Stone
R
, et al
.
Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia
.
J Clin Oncol.
2005
;
23
(
18
):
4110
-
4116
.
40.
Chantepie
SP
,
Reboursiere
E
,
Mear
JB
, et al
.
Gemtuzumab ozogamicin in combination with intensive chemotherapy in relapsed or refractory acute myeloid leukemia
.
Leuk Lymphoma.
2015
;
56
(
8
):
2326
-
2330
.
41.
Prince
HM
,
Kim
YH
,
Horwitz
SM
, et al;
ALCANZA study group
.
Brentuximab vedotin or physician’s choice in CD30-positive cutaneous T-cell lymphoma (ALCANZA): an international, open-label, randomised, phase 3, multicentre trial
.
Lancet.
2017
;
390
(
10094
):
555
-
566
.
42.
Richardson
PG
,
Lee
HC
,
Abdallah
AO
, et al
.
Single-agent belantamab mafodotin for relapsed/refractory multiple myeloma: analysis of the lyophilised presentation cohort from the pivotal DREAMM-2 study
.
Blood Cancer J.
2020
;
10
(
10
):
106
.
43.
Bussel
JB
,
Lee
CS
,
Seery
C
, et al
.
Rituximab and three dexamethasone cycles provide responses similar to splenectomy in women and those with immune thrombocytopenia of less than two years duration
.
Haematologica.
2014
;
99
(
7
):
1264
-
1271
.
44.
Dierickx
D
,
Kentos
A
,
Delannoy
A.
The role of rituximab in adults with warm antibody autoimmune hemolytic anemia
.
Blood.
2015
;
125
(
21
):
3223
-
3229
.
45.
Huth-Kühne
A
,
Baudo
F
,
Collins
P
, et al
.
International recommendations on the diagnosis and treatment of patients with acquired hemophilia A
.
Haematologica.
2009
;
94
(
4
):
566
-
575
.
46.
Scully
M
,
McDonald
V
,
Cavenagh
J
, et al
.
A phase 2 study of the safety and efficacy of rituximab with plasma exchange in acute acquired thrombotic thrombocytopenic purpura
.
Blood.
2011
;
118
(
7
):
1746
-
1753
.
47.
Solomon
SR
,
Sizemore
CA
,
Ridgeway
M
, et al
.
Safety and efficacy of rituximab-based first line treatment of chronic GVHD
.
Bone Marrow Transplant.
2019
;
54
(
8
):
1218
-
1226
.
48.
Rothenberg
ME
,
Klion
AD
,
Roufosse
FE
, et al;
Mepolizumab HES Study Group
.
Treatment of patients with the hypereosinophilic syndrome with mepolizumab
.
N Engl J Med.
2008
;
358
(
12
):
1215
-
1228
.
49.
van Rhee
F
,
Wong
RS
,
Munshi
N
, et al
.
Siltuximab for multicentric Castleman’s disease: a randomised, double-blind, placebo-controlled trial
.
Lancet Oncol.
2014
;
15
(
9
):
966
-
974
.
50.
Locatelli
F
,
Jordan
MB
,
Allen
C
, et al
.
Emapalumab in children with primary hemophagocytic lymphohistiocytosis
.
N Engl J Med.
2020
;
382
(
19
):
1811
-
1822
.
51.
Scott
LJ.
Tocilizumab: a review in rheumatoid arthritis [published correction appears in Drugs. 2018;78(2):285]
.
Drugs
.
2017
;
77
(
17
):
1865
-
1879
.
52.
Le
RQ
,
Li
L
,
Yuan
W
, et al
.
FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome
.
Oncologist.
2018
;
23
(
8
):
943
-
947
.
53.
Salama
C
,
Han
J
,
Yau
L
, et al
.
Tocilizumab in patients hospitalized with Covid-19 pneumonia
.
N Engl J Med.
2021
;
384
(
1
):
20
-
30
.
54.
Turner
AP
,
Knechtle
SJ.
Induction immunosuppression in liver transplantation: a review
.
Transpl Int.
2013
;
26
(
7
):
673
-
683
.
55.
Liu
SN
,
Zhang
XH
,
Xu
LP
, et al
.
Prognostic factors and long-term follow-up of basiliximab for steroid-refractory acute graft-versus-host disease: updated experience from a large-scale study
.
Am J Hematol.
2020
;
95
(
8
):
927
-
936
.
56.
Pardoll
DM.
The blockade of immune checkpoints in cancer immunotherapy
.
Nat Rev Cancer.
2012
;
12
(
4
):
252
-
264
.
57.
Sharma
P
,
Allison
JP.
The future of immune checkpoint therapy
.
Science.
2015
;
348
(
6230
):
56
-
61
.
58.
Wei
SC
,
Duffy
CR
,
Allison
JP.
Fundamental mechanisms of immune checkpoint blockade therapy
.
Cancer Discov.
2018
;
8
(
9
):
1069
-
1086
.
59.
Haanen
J
,
Ernstoff
MS
,
Wang
Y
, et al
.
Autoimmune diseases and immune-checkpoint inhibitors for cancer therapy: review of the literature and personalized risk-based prevention strategy
.
Ann Oncol.
2020
;
31
(
6
):
724
-
744
.
60.
Robert
C
,
Ribas
A
,
Wolchok
JD
, et al
.
Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial
.
Lancet.
2014
;
384
(
9948
):
1109
-
1117
.
61.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
, et al
.
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
.
N Engl J Med.
2012
;
366
(
26
):
2443
-
2454
.
62.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
, et al
.
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science.
2015
;
348
(
6230
):
124
-
128
.
63.
Alexandrov
LB
,
Kim
J
,
Haradhvala
NJ
, et al;
PCAWG Consortium
.
The repertoire of mutational signatures in human cancer
.
Nature.
2020
;
578
(
7793
):
94
-
101
.
64.
Alexandrov
LB
,
Nik-Zainal
S
,
Wedge
DC
, et al;
ICGC PedBrain
.
Signatures of mutational processes in human cancer [published correction appears in Nature. 2013;502(7470):258]
.
Nature
.
2013
;
500
(
7463
):
415
-
421
.
65.
Green
MR
,
Monti
S
,
Rodig
SJ
, et al
.
Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma
.
Blood.
2010
;
116
(
17
):
3268
-
3277
.
66.
Patsoukis
N
,
Wang
Q
,
Strauss
L
,
Boussiotis
VA.
Revisiting the PD-1 pathway
.
Sci Adv.
2020
;
6
(
38
):
eabd2712
.
67.
Ansell
SM
,
Lesokhin
AM
,
Borrello
I
, et al
.
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma
.
N Engl J Med.
2015
;
372
(
4
):
311
-
319
.
68.
Moskowitz
CH
, Ribrag V, Michot JM, et al
.
PD-1 blockade with the monoclonal antibody pembrolizumab (MK-3475) in patients with classical Hodgkin lymphoma after brentuximab vedotin failure: preliminary results from a phase 1b study (KEYNOTE-013)
.
Blood.
2014
;
124
(
21
). Abstract 290.
69.
Dufva
O
,
Pölönen
P
,
Brück
O
, et al
.
Immunogenomic landscape of hematological malignancies [abstract, published correction appears in
Cancer Cell. 2020 38(3):424-428.
Cancer Cell.
2020
;
38
(
3
):
380
-
399
.
70.
Kong
Y
,
Zhu
L
,
Schell
TD
, et al
.
T-cell immunoglobulin and ITIM domain (TIGIT) associates with CD8+ T-cell exhaustion and poor clinical outcome in AML patients
.
Clin Cancer Res.
2016
;
22
(
12
):
3057
-
3066
.
71.
Yang
ZZ
,
Kim
HJ
,
Villasboas
JC
, et al
.
Expression of LAG-3 defines exhaustion of intratumoral PD-1+ T cells and correlates with poor outcome in follicular lymphoma
.
Oncotarget.
2017
;
8
(
37
):
61425
-
61439
.
72.
Luke
JJ
, Patel MR, Hamilton EP, et al
.
A phase I, first-in-human, open-label, dose-escalation study of MGD013, a bispecific DART molecule binding PD-1 and LAG-3, in patients with unresectable or metastatic neoplasms [abstract]
.
J Clin Oncol.
2020
;
38
(
15_suppl
). Abstract 3004.
73.
Williams
P
,
Basu
S
,
Garcia-Manero
G
, et al
.
The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia
.
Cancer.
2019
;
125
(
9
):
1470
-
1481
.
74.
Kikushige
Y
,
Shima
T
,
Takayanagi
S
, et al
.
TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells
.
Cell Stem Cell.
2010
;
7
(
6
):
708
-
717
.
75.
Chao
MP
,
Alizadeh
AA
,
Tang
C
, et al
.
Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia
.
Cancer Res.
2011
;
71
(
4
):
1374
-
1384
.
76.
Chao
MP
,
Alizadeh
AA
,
Tang
C
, et al
.
Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma
.
Cell.
2010
;
142
(
5
):
699
-
713
.
77.
Majeti
R
,
Chao
MP
,
Alizadeh
AA
, et al
.
CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells
.
Cell.
2009
;
138
(
2
):
286
-
299
.
78.
Pang
WW
,
Pluvinage
JV
,
Price
EA
, et al
.
Hematopoietic stem cell and progenitor cell mechanisms in myelodysplastic syndromes
.
Proc Natl Acad Sci USA.
2013
;
110
(
8
):
3011
-
3016
.
79.
Tseng
D
,
Volkmer
JP
,
Willingham
SB
, et al
.
Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response
.
Proc Natl Acad Sci USA.
2013
;
110
(
27
):
11103
-
11108
.
80.
Sallman
DA
, Asch AS, Al Malki MM, et al
.
The first-in-class anti-CD47 antibody magrolimab (5f9) in combination with azacitidine is effective in MDS and AML patients: ongoing phase 1b results [abstract]
.
Blood.
2019
;
134
(
suppl_1
). Abstract 569.
81.
Suntharalingam
G
,
Perry
MR
,
Ward
S
, et al
.
Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412
.
N Engl J Med.
2006
;
355
(
10
):
1018
-
1028
.
82.
Gopal
AK
,
Levy
R
,
Houot
R
, et al
.
First-in-human study of utomilumab, a 4-1BB/CD137 agonist, in combination with rituximab in patients with follicular and other CD20+ non-Hodgkin lymphomas
.
Clin Cancer Res.
2020
;
26
(
11
):
2524
-
2534
.
83.
Ansell
SM
,
Flinn
I
,
Taylor
MH
, et al
.
Safety and activity of varlilumab, a novel and first-in-class agonist anti-CD27 antibody, for hematologic malignancies
.
Blood Adv.
2020
;
4
(
9
):
1917
-
1926
.
84.
Vincenti
F
,
Rostaing
L
,
Grinyo
J
, et al
.
Belatacept and long-term outcomes in kidney transplantation
.
N Engl J Med.
2016
;
374
(
4
):
333
-
343
.
85.
Watkins
B
,
Qayed
M
,
McCracken
C
, et al
.
Phase II trial of costimulation blockade with abatacept for prevention of acute GVHD
.
J Clin Oncol.
2021
;
39
(
17
):
1865
-
1877
.
86.
Watkins
BK
, et al
.
T cell costimulation blockade with CTLA4-Ig (abatacept) for acute Gvhd prevention in HLA matched and mismatched unrelated donor transplantation: results of the first phase 2 trial
.
Biol Blood Marrow Transplant.
2019
;
25
(
3
):
S51
-
S52
.
87.
Tanaka
J
,
Tanaka
N
,
Wang
YH
, et al
.
Phase I study of cellular therapy using ex vivo expanded natural killer cells from autologous peripheral blood mononuclear cells combined with rituximab-containing chemotherapy for relapsed CD20-positive malignant lymphoma patients
.
Haematologica.
2020
;
105
(
4
):
e190
-
e193
.
88.
Mathew
JM
,
H-Voss
J
,
LeFever
A
, et al
.
A phase I clinical trial with ex vivo expanded recipient regulatory T cells in living donor kidney transplants
.
Sci Rep.
2018
;
8
(
1
):
7428
.
89
Tumeh
PC
, Koya RC, Chodon T, et al
.
The impact of ex vivo clinical grade activation protocols on human T-cell phenotype and function for the generation of genetically modified cells for adoptive cell transfer therapy
.
J Immunother.
2010
;
33
:
759
-
768
.
90.
Bollard
CM
,
Gottschalk
S
,
Torrano
V
, et al
.
Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins
.
J Clin Oncol.
2014
;
32
(
8
):
798
-
808
.
91.
Lulla
P
, Naik S, Vasileiou S, et al
.
Clinical effects of administering leukemia-specific donor T cells to patients with AML/MDS after allogeneic transplant
.
Blood.
2021
;
137
(
19
):
2585
-
2597
.
92.
Lulla
PD
,
Tzannou
I
,
Vasileiou
S
, et al
.
The safety and clinical effects of administering a multiantigen-targeted T cell therapy to patients with multiple myeloma
.
Sci Transl Med.
2020
;
12
(
554
):
eaaz3339
.
93.
Shen
RR
,
Pham
CD
,
Wu
M
,
Munson
DJ
,
Aftab
BT.
CD19 chimeric antigen receptor (CAR) engineered epstein-barr virus (EBV) specific T cells—an off-the-shelf, allogeneic CAR T-cell immunotherapy platform
.
Cytotherapy.
2019
;
21
(
5
):
S11
.
94.
Wang
X
,
Wong
CW
,
Urak
R
, et al
.
CMVpp65 vaccine enhances the antitumor efficacy of adoptively transferred CD19-redirected CMV-specific T cells
.
Clin Cancer Res.
2015
;
21
(
13
):
2993
-
3002
.
95.
Romee
R
,
Rosario
M
,
Berrien-Elliott
MM
, et al
.
Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia
.
Sci Transl Med.
2016
;
8
(
357
):
357ra123
.
96.
Foltz
JA
, Berrien-Elliott MM, Neal C, et al
.
Cytokine-induced memory-like (ML) NK cells persist for > 2 months following adoptive transfer into leukemia patients with a MHC-compatible hematopoietic cell transplant (HCT)
.
Blood.
2019
;
134
(
suppl_1
):
1954
-
1954
.
97.
Cooley
S
,
He
F
,
Bachanova
V
, et al
.
First-in-human trial of rhIL-15 and haploidentical natural killer cell therapy for advanced acute myeloid leukemia
.
Blood Adv.
2019
;
3
(
13
):
1970
-
1980
.
98.
Koreth
J
,
Matsuoka
K
,
Kim
HT
, et al
.
Interleukin-2 and regulatory T cells in graft-versus-host disease
.
N Engl J Med.
2011
;
365
(
22
):
2055
-
2066
.
99.
Whangbo
JS
,
Kim
HT
,
Mirkovic
N
, et al
.
Dose-escalated interleukin-2 therapy for refractory chronic graft-versus-host disease in adults and children
.
Blood Adv.
2019
;
3
(
17
):
2550
-
2561
.
100.
Sockolosky
JT
,
Trotta
E
,
Parisi
G
, et al
.
Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes
.
Science.
2018
;
359
(
6379
):
1037
-
1042
.
101.
Charych
D
,
Khalili
S
,
Dixit
V
, et al
.
Modeling the receptor pharmacology, pharmacokinetics, and pharmacodynamics of NKTR-214, a kinetically-controlled interleukin-2 (IL2) receptor agonist for cancer immunotherapy
.
PLoS One.
2017
;
12
(
7
):
e0179431
.
102.
Klein
C
,
Waldhauer
I
,
Nicolini
VG
, et al
.
Cergutuzumab amunaleukin (CEA-IL2v), a CEA-targeted IL-2 variant-based immunocytokine for combination cancer immunotherapy: Overcoming limitations of aldesleukin and conventional IL-2-based immunocytokines
.
OncoImmunology.
2017
;
6
(
3
):
e1277306
.
103.
Boyman
O
,
Kovar
M
,
Rubinstein
MP
,
Surh
CD
,
Sprent
J.
Selective stimulation of T cell subsets with antibody-cytokine immune complexes
.
Science.
2006
;
311
(
5769
):
1924
-
1927
.
104.
Levin
AM
,
Bates
DL
,
Ring
AM
, et al
.
Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’
.
Nature.
2012
;
484
(
7395
):
529
-
533
.
105.
Timmerman
JM
,
Byrd
JC
,
Andorsky
DJ
, et al
.
A phase I dose-finding trial of recombinant interleukin-21 and rituximab in relapsed and refractory low grade B-cell lymphoproliferative disorders
.
Clin Cancer Res.
2012
;
18
(
20
):
5752
-
5760
.
106.
Dufner
V
,
Sayehli
CM
,
Chatterjee
M
, et al
.
Long-term outcome of patients with relapsed/refractory B-cell non-Hodgkin lymphoma treated with blinatumomab
.
Blood Adv.
2019
;
3
(
16
):
2491
-
2498
.
107.
Viardot
A
,
Goebeler
ME
,
Hess
G
, et al
.
Phase 2 study of the bispecific T-cell engager (BiTE) antibody blinatumomab in relapsed/refractory diffuse large B-cell lymphoma
.
Blood.
2016
;
127
(
11
):
1410
-
1416
.
108.
Gökbuget
N
,
Dombret
H
,
Bonifacio
M
, et al
.
Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia [published correction appears in Blood. 2019;133(24):2625]
.
Blood
.
2018
;
131
(
14
):
1522
-
1531
.
109.
Topp
MS
,
Gökbuget
N
,
Stein
AS
, et al
.
Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study
.
Lancet Oncol.
2015
;
16
(
1
):
57
-
66
.
110.
Schwartz
M
, Damon LE, Jeyakumar D, et al
.
Blinatumomab in combination with pembrolizumab is safe for adults with relapsed or refractory B-lineage acute lymphoblastic leukemia: University of California Hematologic Malignancies Consortium Study 1504 [abstract]
.
Blood.
2019
;
134
(
suppl_1
). Abstract 3880.
111.
Liu
L
,
Lam
CK
,
Long
V
, et al
.
MGD011, A CD19 x CD3 dual-affinity retargeting bi-specific molecule incorporating extended circulating half-life for the treatment of B-cell malignancies
.
Clin Cancer Res.
2017
;
23
(
6
):
1506
-
1518
.
112.
Reusch
U
,
Duell
J
,
Ellwanger
K
, et al
.
A tetravalent bispecific TandAb (CD19/CD3), AFM11, efficiently recruits T cells for the potent lysis of CD19(+) tumor cells
.
MAbs.
2015
;
7
(
3
):
584
-
604
.
113.
Schuster
SJ
, Bartlett NL, Assouline S, et al
.
Mosunetuzumab induces complete remissions in poor prognosis non-Hodgkin lymphoma patients, including those who are resistant to or relapsing after chimeric antigen receptor T-cell (CAR-T) therapies, and is active in treatment through multiple lines [abstract]
.
Blood.
2019
;
134
(
suppl_1
). Abstract 6.
114.
Rothe
A
,
Sasse
S
,
Topp
MS
, et al
.
A phase 1 study of the bispecific anti-CD30/CD16A antibody construct AFM13 in patients with relapsed or refractory Hodgkin lymphoma
.
Blood.
2015
;
125
(
26
):
4024
-
4031
.
115.
Uy
GL
, Godwin J, Rettig MP, et al
.
Preliminary results of a phase 1 study of flotetuzumab, a CD123 x CD3 bispecific Dart® protein, in patients with relapsed/refractory acute myeloid leukemia and myelodysplastic syndrome [abstract]
.
Blood.
2017
;
130
(
suppl_1
). Abstract 637.
116.
Jabbour
E
,
Düll
J
,
Yilmaz
M
, et al
.
Outcome of patients with relapsed/refractory acute lymphoblastic leukemia after blinatumomab failure: no change in the level of CD19 expression
.
Am J Hematol.
2018
;
93
(
3
):
371
-
374
.
117.
Topp
MS
,
Gökbuget
N
,
Zugmaier
G
, et al
.
Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia
.
J Clin Oncol.
2014
;
32
(
36
):
4134
-
4140
.
118.
Duell
J
,
Lammers
PE
,
Djuretic
I
, et al
.
Bispecific antibodies in the treatment of hematologic malignancies
.
Clin Pharmacol Ther.
2019
;
106
(
4
):
781
-
791
.
119.
Legut
M
,
Dolton
G
,
Mian
AA
,
Ottmann
OG
,
Sewell
AK.
CRISPR-mediated TCR replacement generates superior anticancer transgenic T cells
.
Blood.
2018
;
131
(
3
):
311
-
322
.
120.
Mastaglio
S
,
Genovese
P
,
Magnani
Z
, et al
.
NY-ESO-1 TCR single edited stem and central memory T cells to treat multiple myeloma without graft-versus-host disease
.
Blood.
2017
;
130
(
5
):
606
-
618
.
121.
Maude
SL
,
Laetsch
TW
,
Buechner
J
, et al
.
Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia
.
N Engl J Med.
2018
;
378
(
5
):
439
-
448
.
122.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
, et al
.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med.
2017
;
377
(
26
):
2531
-
2544
.
123.
Schuster
SJ
,
Bishop
MR
,
Tam
CS
, et al;
JULIET Investigators
.
Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma
.
N Engl J Med.
2019
;
380
(
1
):
45
-
56
.
124.
Wang
M
,
Munoz
J
,
Goy
A
, et al
.
KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma
.
N Engl J Med.
2020
;
382
(
14
):
1331
-
1342
.
125.
FDA approves lisocabtagene maraleucel for relapsed or refractory large B-cell lymphoma
. https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-lisocabtagene-maraleucel-relapsed-or-refractory-large-b-cell-lymphoma (
2021
).
126.
Munshi
NC
,
Anderson
LD
Jr
,
Shah
N
, et al
.
Idecabtagene vicleucel in relapsed and refractory multiple myeloma
.
N Engl J Med.
2021
;
384
(
8
):
705
-
716
.
127.
Zhao
L
,
Cao
YJ.
Engineered T cell therapy for cancer in the clinic
.
Front Immunol.
2019
;
10
:
2250
.
128.
Rafiq
S
,
Hackett
CS
,
Brentjens
RJ.
Engineering strategies to overcome the current roadblocks in CAR T cell therapy
.
Nat Rev Clin Oncol.
2020
;
17
(
3
):
147
-
167
.
129.
Shah
NN
,
Fry
TJ.
Mechanisms of resistance to CAR T cell therapy
.
Nat Rev Clin Oncol.
2019
;
16
(
6
):
372
-
385
.
130.
Brudno
JN
,
Kochenderfer
JN.
Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management
.
Blood Rev.
2019
;
34
:
45
-
55
.
131.
Kochenderfer
JN
,
Wilson
WH
,
Janik
JE
, et al
.
Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19
.
Blood.
2010
;
116
(
20
):
4099
-
4102
.
132.
Porter
DL
, Hwang W, Frey NV, et al
.
Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia
.
Sci Transl Med.
2015
;
7
:
303ra139
.
133.
Fraietta
JA
,
Lacey
SF
,
Orlando
EJ
, et al
.
Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia
.
Nat Med.
2018
;
24
(
5
):
563
-
571
.
134.
Choi
BD
,
Yu
X
,
Castano
AP
, et al
.
CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity
.
Nat Biotechnol.
2019
;
37
(
9
):
1049
-
1058
.
135.
Shah
NN
,
Johnson
BD
,
Schneider
D
, et al
.
Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial
.
Nat Med.
2020
;
26
(
10
):
1569
-
1575
.
136.
Qasim
W
,
Zhan
H
,
Samarasinghe
S
, et al
.
Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells
.
Sci Transl Med.
2017
;
9
(
374
):
eaaj2013
.
137.
Stadtmauer
EA
,
Fraietta
JA
,
Davis
MM
, et al
.
CRISPR-engineered T cells in patients with refractory cancer
.
Science.
2020
;
367
(
6481
):
eaba7365
.
138.
Rupp
LJ
,
Schumann
K
,
Roybal
KT
, et al
.
CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells
.
Sci Rep.
2017
;
7
(
1
):
737
.
139.
Hirakawa
MP
,
Krishnakumar
R
,
Timlin
JA
,
Carney
JP
,
Butler
KS.
Gene editing and CRISPR in the clinic: current and future perspectives
.
Biosci Rep.
2020
;
40
(
4
):
BSR20200127
.
140.
Li
AM
, Hucks GE, Dinofia AM, et al
.
Checkpoint inhibitors augment CD19-directed chimeric antigen receptor (CAR) T cell therapy in relapsed B-cell acute lymphoblastic leukemia [abstract]
.
Blood.
2018
;
132
(
suppl 1
). Abstract 556.
141.
Kueberuwa
G
,
Kalaitsidou
M
,
Cheadle
E
,
Hawkins
RE
,
Gilham
DE.
CD19 CAR T cells expressing IL-12 eradicate lymphoma in fully lymphoreplete mice through induction of host immunity
.
Mol Ther Oncolytics.
2017
;
8
:
41
-
51
.
142.
Yeku
OO
,
Purdon
TJ
,
Koneru
M
,
Spriggs
D
,
Brentjens
RJ.
Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment
.
Sci Rep.
2017
;
7
(
1
):
10541
.
143.
Hurton
LV
,
Singh
H
,
Najjar
AM
, et al
.
Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells
.
Proc Natl Acad Sci USA.
2016
;
113
(
48
):
E7788
-
E7797
.
144.
Oldenburg
J
,
Mahlangu
JN
,
Kim
B
, et al
.
Emicizumab prophylaxis in hemophilia A with inhibitors
.
N Engl J Med.
2017
;
377
(
9
):
809
-
818
.
145.
Elinav
E
,
Waks
T
,
Eshhar
Z.
Redirection of regulatory T cells with predetermined specificity for the treatment of experimental colitis in mice
.
Gastroenterology.
2008
;
134
(
7
):
2014
-
2024
.
146.
Fransson
M
,
Piras
E
,
Burman
J
, et al
.
CAR/FoxP3-engineered T regulatory cells target the CNS and suppress EAE upon intranasal delivery
.
J Neuroinflammation.
2012
;
9
(
1
):
112
.
147.
Zhang
Q
,
Lu
W
,
Liang
CL
, et al
.
Chimeric antigen receptor (CAR) Treg: a promising approach to inducing immunological tolerance
.
Front Immunol.
2018
;
9
:
2359
.
148.
Boardman
DA
,
Philippeos
C
,
Fruhwirth
GO
, et al
.
Expression of a chimeric antigen receptor specific for donor HLA class I enhances the potency of human regulatory T cells in preventing human skin transplant rejection
.
Am J Transplant.
2017
;
17
(
4
):
931
-
943
.
149.
Noyan
F
,
Zimmermann
K
,
Hardtke-Wolenski
M
, et al
.
Prevention of allograft rejection by use of regulatory T cells with an MHC-specific chimeric antigen receptor
.
Am J Transplant.
2017
;
17
(
4
):
917
-
930
.
150.
Ellebrecht
CT
,
Bhoj
VG
,
Nace
A
, et al
.
Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease
.
Science.
2016
;
353
(
6295
):
179
-
184
.
151.
Rodrigues
CMC
,
Plotkin
SA.
Impact of vaccines; health, economic and social perspectives
.
Front Microbiol.
2020
;
11
:
1526
.
152.
Baden
LR
, El Sahly HM, Essink B, et al
.
Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine
.
N Engl J Med.
2021
;
384
:
403
-
416
.
153.
Boehm
BE
,
Cornell
JE
,
Wang
H
,
Mukherjee
N
,
Oppenheimer
JS
,
Svatek
RS.
Efficacy of Bacillus Calmette-Guérin strains for treatment of nonmuscle invasive bladder cancer: a systematic review and network meta-analysis
.
J Urol.
2017
;
198
(
3
):
503
-
510
.
154.
Cheever
MA
,
Higano
CS.
PROVENGE (Sipuleucel-T) in prostate cancer: the first FDA-approved therapeutic cancer vaccine
.
Clin Cancer Res.
2011
;
17
(
11
):
3520
-
3526
.
155.
Koehne
G
, Devlin S, Chung DJ, et al
.
WT1 heteroclitic epitope immunization following autologous stem cell transplantation in patients with high-risk multiple myeloma (MM) [abstract]
.
J Clin Oncol.
2017
;
35
(
15_suppl
). Abstract 8016.
156.
Hailemichael
Y
,
Dai
Z
,
Jaffarzad
N
, et al
.
Persistent antigen at vaccination sites induces tumor-specific CD8+ T cell sequestration, dysfunction and deletion
.
Nat Med.
2013
;
19
(
4
):
465
-
472
.
157.
Antonios
JP
,
Soto
H
,
Everson
RG
, et al
.
PD-1 blockade enhances the vaccination-induced immune response in glioma
.
JCI Insight.
2016
;
1
(
10
):
e87059
.
158.
Soares
KC
,
Rucki
AA
,
Wu
AA
, et al
.
PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors
.
J Immunother.
2015
;
38
(
1
):
1
-
11
.
159.
Wilgenhof
S
,
Corthals
J
,
Heirman
C
, et al
.
Phase II study of autologous monocyte-derived mRNA electroporated dendritic cells (TriMixDC-MEL) plus ipilimumab in patients with pretreated advanced melanoma
.
J Clin Oncol.
2016
;
34
(
12
):
1330
-
1338
.
160.
Hodi
FS
,
O’Day
SJ
,
McDermott
DF
, et al
.
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med.
2010
;
363
(
8
):
711
-
723
.
161.
Wu
AA
,
Bever
KM
,
Ho
WJ
, et al
.
A phase II study of allogeneic GM-CSF-transfected pancreatic tumor vaccine (GVAX) with ipilimumab as maintenance treatment for metastatic pancreatic cancer
.
Clin Cancer Res.
2020
;
26
(
19
):
5129
-
5139
.
162.
Hollingsworth
RE
,
Jansen
K.
Turning the corner on therapeutic cancer vaccines
.
NPJ Vaccines.
2019
;
4
(
1
):
7
.
163.
Kaufman
HL
,
Kohlhapp
FJ
,
Zloza
A.
Oncolytic viruses: a new class of immunotherapy drugs [published correction appears in Nat Rev Drug Discov. 2016;15(9):660]
.
Nat Rev Drug Discov
.
2015
;
14
(
9
):
642
-
662
.
164.
Bommareddy
PK
,
Patel
A
,
Hossain
S
,
Kaufman
HL.
Talimogene laherparepvec (T-VEC) and other oncolytic viruses for the treatment of melanoma
.
Am J Clin Dermatol.
2017
;
18
(
1
):
1
-
15
.
165.
Marshall
E.
Gene therapy death prompts review of adenovirus vector
.
Science.
1999
;
286
(
5448
):
2244
-
2245
.
166.
Raja
J
,
Ludwig
JM
,
Gettinger
SN
,
Schalper
KA
,
Kim
HS.
Oncolytic virus immunotherapy: future prospects for oncology
.
J Immunother Cancer.
2018
;
6
(
1
):
140
.
167.
Bauerschmitz
GJ
,
Kanerva
A
,
Wang
M
, et al
.
Evaluation of a selectively oncolytic adenovirus for local and systemic treatment of cervical cancer
.
Int J Cancer.
2004
;
111
(
2
):
303
-
309
.
168.
Roberts
DM
,
Nanda
A
,
Havenga
MJ
, et al
.
Hexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity
.
Nature.
2006
;
441
(
7090
):
239
-
243
.
169.
Zaiss
AK
,
Machado
HB
,
Herschman
HR.
The influence of innate and pre-existing immunity on adenovirus therapy
.
J Cell Biochem.
2009
;
108
(
4
):
778
-
790
.
170.
Park
AK
,
Fong
Y
,
Kim
SI
, et al
.
Effective combination immunotherapy using oncolytic viruses to deliver CAR targets to solid tumors
.
Sci Transl Med.
2020
;
12
(
559
):
eaaz1863
.
171.
Hemminki
O
,
Dos Santos
JM
,
Hemminki
A.
Oncolytic viruses for cancer immunotherapy
.
J Hematol Oncol.
2020
;
13
(
1
):
84
.
172.
Park
JH
,
Rivière
I
,
Gonen
M
, et al
.
Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia
.
N Engl J Med.
2018
;
378
(
5
):
449
-
459
.
173.
Zhang
X
,
Chen
BD
,
Zhao
LD
,
Li
H.
The gut microbiota: emerging evidence in autoimmune diseases
.
Trends Mol Med.
2020
;
26
(
9
):
862
-
873
.
174.
Zitvogel
L
,
Ma
Y
,
Raoult
D
,
Kroemer
G
,
Gajewski
TF.
The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies
.
Science.
2018
;
359
(
6382
):
1366
-
1370
.
175.
Gopalakrishnan
V
,
Spencer
CN
,
Nezi
L
, et al
.
Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients
.
Science.
2018
;
359
(
6371
):
97
-
103
.
176.
Frey
N
,
Porter
D.
Cytokine release syndrome with chimeric antigen receptor T cell therapy
.
Biol Blood Marrow Transplant.
2019
;
25
(
4
):
e123
-
e127
.
177.
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
(
9967
):
517
-
528
.
178.
Tawbi
HA
,
Forsyth
PA
,
Algazi
A
, et al
.
Combined nivolumab and ipilimumab in melanoma metastatic to the brain
.
N Engl J Med.
2018
;
379
(
8
):
722
-
730
.
179.
Wang
DY
,
Salem
JE
,
Cohen
JV
, et al
.
Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis
.
JAMA Oncol.
2018
;
4
(
12
):
1721
-
1728
.
180.
Liu
E
,
Marin
D
,
Banerjee
P
, et al
.
Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors
.
N Engl J Med.
2020
;
382
(
6
):
545
-
553
.
181.
Silva
DA
,
Yu
S
,
Ulge
UY
, et al
.
De novo design of potent and selective mimics of IL-2 and IL-15
.
Nature.
2019
;
565
(
7738
):
186
-
191
.
182.
Eckl
J
,
Raffegerst
S
,
Schnorfeil
F
, et al
.
DC vaccination induces antigen specific immune responses in AML patients: a 1-year interim assessment [abstract]
.
Blood.
2019
;
134
(
suppl_1
). Abstract 3923.