We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
ReviewOpen Accesscc iconby iconnc iconnd icon

Immunotherapy in hematological malignancies: recent advances and open questions

    Essam A Tawfik

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    National Center for Pharmaceutical Technology, Life Science & Environment Research Institute, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Norah A Aldrak

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Shahad H Albrahim

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Dunia A Alzahrani

    National Center for Pharmaceutical Technology, Life Science & Environment Research Institute, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Haya A Alfassam

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Samar M Alkoblan

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Abdulaziz M Almalik

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    National Center for Pharmaceutical Technology, Life Science & Environment Research Institute, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Kok-Siong Chen

    BWH Center of Excellence for Biomedicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA

    Center for Stem Cell & Translational Immunotherapy, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA

    ,
    Rana Abou-Khalil

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    ,
    Khalid Shah

    BWH Center of Excellence for Biomedicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA

    Center for Stem Cell & Translational Immunotherapy, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA

    Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA

    &
    Nada M Zaidan

    *Author for correspondence:

    E-mail Address: nzaidan@kacst.edu.sa

    Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia

    Published Online:https://doi.org/10.2217/imt-2021-0065

    Abstract

    Over recent years, tremendous advances in immunotherapy approaches have been observed, generating significant clinical progress. Cancer immunotherapy has been shown, in different types of blood cancers, to improve the overall survival of patients. Immunotherapy treatment of hematopoietic malignancies is a newly growing field that has been accelerating over the past years. Several US FDA approved drugs and cell-based therapies are being exploited in the late stage of clinical trials. This review attempt to highlight and discuss the numerous innovative immunotherapy approaches of hematopoietic malignancy ranging from nonmyeloablative transplantation, T-cell immunotherapy, natural killer cells and immune agonist to monoclonal antibodies and vaccination. In addition, a brief discussion on the future advances and accomplishments required to counterpart the current immunotherapeutic approaches for hematopoietic malignancies were also highlighted.

    Graphical abstract

    Hematopoiesis is a highly dynamic regeneration process giving rise to all types of mammalian blood cells during embryonic development and adulthood [1]. It requires both self-renewal of hematopoietic stem cells (HSCs), that reside within the bone marrow niche, and a hierarchical cascade of progenitor differentiation [2]. HSCs are defined by their capability to differentiate into immature progenitor cells, i.e. myeloid cells (myeloid lineage) and lymphoid cells (lymphoid lineage) [2]. The myeloid cells produce platelets, erythrocytes, granulocytes, monocytes, macrophages and dendritic cells, whereas the lymphoid cells give rise to B and T lymphocytes, plasma cells and natural killer (NK) cells [3], as shown in Figure 1.

    Figure 1. Hematopoiesis: stages of blood cell development within the bone marrow niche.

    Created with Biorender.com.

    Hematopoiesis is governed by a combination of extrinsic and intrinsic factors, such as niche-associated factors, transcription factors, signal transduction pathways and chromatin modifiers. Any disruption of these factors may lead to serious hematological disorders, including hematologic malignancies, which can occur at any stage of blood cell differentiation; hence, affecting the production of blood cells. Consequently, HSCs would lose their functions of infection resistance and hemostasis [4]. Hematopoietic transformation can be resulted from the disruption of the transcription factors, including RUNX1/AML1, GATA3 and ETS that determine normal hematopoiesis [4,5]. Several studies have explored the relationship between the silencing of chromatin-modifying enzymes or misregulation of transcription factors and the development of hematological malignancies through epigenetic alterations [6]. Therefore, inhibiting the chromatin modifiers or reversing the transcription misregulation may rectify these epigenetic alterations, and thereby, procure treatments for these malignancies [7].

    The microenvironment of hematologic malignancies consists of bone marrow (BM), blood vessels and peripheral lymphoid organs that provide nutrition and survival factors. Soluble factors such as VEGF, BFGF, IL-3, IL-6, nitric oxide (NO), APRIL and many others, could stimulate tumor cell survival and growth [8].

    Bone marrow niche includes HSCs and nonhematopoietic cells, in other words, endothelial cells, fibroblasts, osteoblasts, macrophages, mast cells and mesenchymal stem cells (MSCs) [9]. Genetic alterations in the BM niche cells, in other words, HSCs or hematopoietic progenitor cells (HPCs) can modify the microenvironment and the proliferation of malignancies [10–12]. Any alterations in the signaling pathways can be a trigger to developing hematopoietic malignancies (Figure 2). In addition, the reduction of hematopoietic cells or HSCs can also enhance the progression of hematopoietic malignancies [13], by inhibiting the differentiation of HSCs into HPCs [14].

    Figure 2. The transformation process of the normal hematopoietic niche into leukemic niche including changes in the signaling pathways, such a TGFβ and Notch.

    Within the leukemic niche, factors that promote HSCs will decrease, while factors that stimulates leukemic cells will increase. Created with Biorender.com.

    HSC: Hematopoietic stem cell.

    It is not only the niche that supports the survival of cancer cells at the expense of HSCs, hematological neoplasms can also hijack normal bone marrow to secrete high levels of stem cell factor (SCF), in order to attract normal CD34+ migrating into the cancer niche [15]. Abnormalities in the level of cytokines, such as IL-6, G-CSF, and TNF-α; CCL3, and CCL4; and the imbalance between suppressor and potentiator genes in the bone marrow microenvironment, could play important roles in the development of hematopoietic malignancies.

    These microenvironment alterations contribute to the formation of a malignancy-favorable environment for supporting cancer survival throughout niche-associated factors. The cellular components of the bone marrow and lymph node niche, and the means by which they modulate normal and malignant hematopoiesis have been widely investigated in the past decade. Targeting malignant microenvironments, such as the niche remodeling; or the suppression of normal hematopoiesis, could be valuable approaches to treat hematopoietic cancers, overcome chemotherapeutic resistance and develop novel antineoplastic agents.

    In this review, we highlight the recent immunomodulatory therapeutic approaches that are currently employed for the treatment of hematopoietic malignancies.

    Hematopoietic cell transplantation

    In the past two decades, hematopoietic cell transplantation was used for the treatment of several types of cancers including hematological cancers [16,17]. There are three known approaches that are used as cancer treatment regimens, in other words, the nonmyeloablative transplantation, also known as nonmyeloablative conditioning (NMA), myeloablative conditioning (MA) and reduced-intensity conditioning (RIC). MA requires a stem cell support, owing to the irreversible reduction in mature blood cells (cytopenia). On the other hand, patients who undergo NMA do not require stem cell support, as it is less likely to develop cytopenia. RIC does not fit the criteria of the previous two conditioning, however, stem cell support is crucial with this approach, with a possibility of reversible cytopenia [18,19]. Therefore, only hematopoietic cell transplantation approach NMA will be discussed as a hematological malignancy treatment regimen.

    The nonmyeloablative transplantation relies on the use of immune cells of a donor to destroy the cancer of a recipient individual. Therefore, NMA is the most commonly used hematopoietic approach, in which it can reduce the toxicity of other cancer therapies, such as chemo- and radiotherapies [20]. NMA is being exploited as a timing regimen that emphasizes the immunosuppression, decreases the risk of rejection by residual recipient immunity, and produces mixed chimerism [21]. To determine the chimerism in the recipient blood samples, quantification of the percentage of hematopoietic cell recovery from donors or recipients origins can be performed. Mixed chimerism describes a state when both the recipient and donor hematopoietic stem cells coexist [22].

    Among other studies, a multicenter 10-year retrospective study involved 70 allogeneic hematopoietic stem cell transplantation (HSCT), including 33 patients with mantle cell lymphoma, allowing better progression-free survival (PFS) rates when treated with NMA [23]. A different study with a prospective Phase II trial has used NMA with myeloma patients and the findings suggested that NMA regimen brought clinical advantages to the patients [24]. Another study was also evaluating NMA on lymphoma patients reporting that NMA could enhance the overall survival rate [25].

    Nevertheless, there are number of cases that have demonstrated a graft-versus-host disease (GvHD) and progression or relapse of the treated cancer after the use of NMA [26]. In addition, despite of the promising results of the clinical studies using NMA in hematological malignances, more than 50% of those studies have no published results, which would require more considerations in the near future [27]. Consequently, this treatment approach is directed to the infirm patients, who might be inclined to other diseases. Hence, NMA can be used to decrease the risk of cancer therapy-related morbidity and to cure vicious cancers, such as hematopoietic malignancies [28].

    T-cell immunotherapy

    The use of T cells as a potential approach for immunotherapy has gained major recognition in the past two decades [29]. T cells have the potential to recognize diverse antigens from pathogens, tumors and the environment, while maintaining immunological memory and self-tolerance [30]. T-cell immunotherapy, or adoptive T-cell (ATC) therapy, is the process of infusing autologous T cells into cancer patients for treatment purposes [31]. An important question in assessing ATC therapeutic strategies is whether T-cell therapy has the capacity to recognize tumor-associated antigens (TAAs) [32]. Although immune cells are recruited to the tumor microenvironment, they display diminished anti-tumor functions due to immunosuppression from tumor-derived signals [33]. The aim of ATC therapy is to overcome the tumor-induced immunosuppression by ex vivo activation and expansion of cancer-specific T cells, and then reinfusion for subsequent killing upon recognizing tumor antigens [34]. In this section, we will discuss the efforts taken to develop tumor-specific T cells for ATC therapy in the context of hematological malignancies.

    Marrow infiltrating lymphocytes

    Marrow-infiltrating lymphocytes (MILs) are likely to provide similar benefits to tumor-infiltrating lymphocytes (TILs) as they possess antigenic specificity for hematological malignancies [35]. It has been observed that antigen-specific memory T cells are enriched in the bone marrow and are involved in controlling the tumor dormancy, providing therapeutic activity [36]. As opposed to TILs, MILs exists in all patients, and therefore, can be easily harvested and rapidly expanded [37]. Ex vivo activation and expansion of MILs have demonstrated significant antitumor specificities for mature plasma cells and their clonogenic precursors in multiple myeloma compared with their peripheral blood counterparts [38]. The first clinical study using MILs for ATC therapy showed a direct correlation between their tumor specificity and clinical response in multiple myeloma, as well as, the persistence of tumor-specific T cells in the bone marrow [37].

    Chimeric antigen receptor T cells

    Challenges associated with the ex vivo expansion of tumor-specific T cells have led to the development of engineered T cells, however, the response of these cells was limited to tumor antigens presented by major histocompatibility complex (MHC) [31]. The genetic engineering of T cells to have antitumor specificities could allow for rapid production of tumor-specific T cells for therapeutic applications [29]. Chimeric antigen receptors (CARs) are synthetic receptors that enable T cells to recognize and eliminate cells, expressing their cognate ligands [39]. Patients autologous T cells are isolated and genetically modified to express a CAR that is specific to TAA, then expanded and infused into the same patient [31]. T cells expressing CARs allow for combining their cytotoxic functions with a high degree of antibody specificity by recognizing specific TAAs in a MHC-independent manner [40]. This ability is particularly advantageous, as the loss of MHC is a common immune evasion mechanism of malignant cells to escape T-cell recognition [41].

    Over the past decade, CAR T-cell therapy has been one of the most outstanding therapeutic advances for certain hematopoietic malignancies, such as leukemia and B cell lymphoma [42,43]. Initial clinical trials have used CAR T cells directed against CD19, expressed by a wide range of B cells (pro to memory B cells) [44]. CD19 is known to be expressed in various B cell malignancies, such as acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), and B cell lymphoma, and its absence in other hematopoietic cancers makes it an ideal CAR target [45]. In 2010, Kochenderfer et al. reported on dramatic regression of advanced follicular lymphoma using anti-CD19 CAR T cells [46]. For 39 weeks post CAR T-cell infusion, B cells were absent, while other blood counts were promptly recovered [46]. Another study included 15 patients suffering from diffuse large B cell lymphoma, indolent lymphoma or CLL. It was reported that eight out of 15 patients have achieved a complete remission (CR), while four patients have achieved a partial remission (PR), after receiving a single infusion of anti-CD19 CAR T cells following conditioning chemotherapy [47]. Brentjens et al. have reported rapid tumor eradication and achievement of CR in adult patients with refractory B cell ALL (B-ALL) [48]. It has also been reported that anti-CD19 CAR T-cell therapy is therapeutically effective in high-risk B-ALL patients, including those with Philadelphia chromosome-positive disease that occur in relapsed B-ALL patients who undergone allogeneic hematopoietic stem cell transplant [49]. In a trial aimed at treating relapsed/refractory (R/R) CLL, the overall response was 57%, with approximately 29% CR, 4+ years of CAR T-cell persistence and no relapse [50].

    CAR T cells have demonstrated great success in treating certain hematological malignancies [31]. However, this therapeutic approach face certain limitations on the path of development [51]. Upon CAR T-cell activation, excessive cytokine production can potentially lead to cytokine toxicity, or cytokine release syndrome, which could be a potential life-threatening condition [52]. Efforts are being made to define diagnostic criteria for severe cytokine release syndrome, where therapeutic intervention is required to minimize the associated symptom [49]. Another treatment-related toxicity includes on-target off-tumor toxicity. It was reported that anti-CD19 CAR T cells may lead to B-cell aplasia and hypogammaglobinemia, due to off-tumor targeting of CD19 on normal B-cell progenitors [45]. Both effects are well tolerated and manageable by administering immunoglobulins intravenously [39]. Other obstacles include the downregulation of target antigens or tumor antigen heterogeneity that can lead to tumor relapse, a commonly seen phenomenon in CD19+ B-cell malignancies [53]. Several efforts to overcome these drawbacks were recently investigated, such as utilizing more than one CAR T cells targeting different antigens [54] or using bispecific CAR T cells [55]. In the upcoming years, advances in gene editing and the development of innovative techniques to manipulate the immune system will guide this new era of cell-based therapeutics, enhancing the rate of cures from hematological malignancies [43].

    Natural killer cell immunotherapy

    NK cells are cytokine-secreting cytolytic subset of innate lymphoid cells that can induce death in virally-infected or tumor cells [56]. They constitute of approximately 5–15% of circulating lymphocytes, and therefore, represent one of the three major lymphocyte lineages including T and B lymphocytes [57]. As opposed to adaptive lymphocytes, NK cells are involved in innate immune response [58]. One of the key structural features of NK cells is the presence of cytoplasmic granules that contain proteases and perforins [59]. NK cells can deliver immediate response upon recognizing stress and ‘danger’ signals, from foreign molecules [60]. Additionally, NK cells play an important role in modulating immune responses via rapid production of various cytokines and chemokines, eradication of antigen-presenting cells and activation of T cells [57].

    NK cells can attack tumor cells by downregulating HLA class I molecules via ‘missing self-recognition’ or overexpression of ligands for NK activating receptors as ‘induced self-recognition’ [61]. NK cells are recruited to the tumor microenvironment under the direction of pro-inflammatory chemokines secreted from innate and adaptive immune cells [62]. Normally, NK activation is inhibited by the interaction of their inhibitory receptors with MHC class I molecules. However, many cancer cells downregulate their MHC class I expression to avoid CD8+ T cells [63]. Therefore, NK cells possess therapeutic potential for cancer, where other immune cells fail to recognize and eliminate cancer cells due to MHC class I downregulation [64].

    Due to certain limitations in CAR T-cell therapy, including the high costs associated with manufacturing the drug, and its patient-specificity, CAR NK cells represent an appealing solution [61]. Allogenic CAR NK cells can be used without causing GvHD, and thus represent a potential system for generating ‘off-the-shelf’ or ‘universal’ therapeutic cells [65]. Several sources of CAR NK cells occur and have been used in preclinical and clinical settings, including cell lines, such as NK-92, UCB and hematopoietic pluripotent stem cells [65–67]. Numerous clinical trials involving CAR NK cells are targeting CD19, CD22, B-cell maturation antigen (BCMA), CD33 or CD7 for hematological malignancies [68]. In early 2020, Liu et al. published a large-scale CAR NK cell trial (NCT03056339), where NK cells were engineered with an anti-CD19 CAR for treatment of R/R CD19+ cancers [69]. In the Phase I and II trials, eight out of the 11 participants had objective response, including seven that achieved CR. Remarkably, those eight patients showed response to the treatment within the first month postinfusion.

    However, similar to CAR T cells, there are inevitable limitations, from loss of target antigens, to tumor heterogeneity and suppressive tumor microenvironment [68]. Overall, NK cell therapy is evolving as a promising area of clinical research, with manageable safety and preliminary signs of efficacy in patients with certain hematological malignancies [64]. The results of clinical trials that involve alloreactive NK cells in patients with hematological malignancies are encouraging [70]. Further clinical studies with larger patient cohorts are required to validate these promising early results [64].

    Monoclonal antibodies

    Another mechanism of NK cell-mediated elimination of cancer cells is via antibody-dependent cellular cytotoxicity (ADCC). This approach involves the administration of monoclonal antibodies (mAb) directed against TAAs that leads to the recognition and killing of cancer cells [64]. An example of therapeutic mAb in the context of hematological malignancies includes rituximab, an anti-CD20 used for the treatment of lymphoma. Rituximab binds to CD20 antigens that expressed on the surface of immune system B cells, subsequently eradicating the cancerous cells [71]. Several other new US FDA approved drugs using mAb targeting CD20 have been employed to treat hematological malignancies, in other words, ofatumumab and obinutuzumab [72,73]. Ofatumumab consist of a humanized monoclonal antibody that interact with a different CD20 epitope than rituximab, therefore, inducing a better ADCC [72]. The use of obinutuzumab has also shown a greater ADCC and antitumor effect [73].

    In addition to CD20, other FDA approved drugs were used to target other antigens expressed by malignant cells. A study by de Weers et al. has shown that the administration of daratumumab, a mAb targeting CD38 antigen, prompts the killing of multiple myeloma and other hematological tumors [74]. Moreover, brentuximab vedotin, an antibody drug conjugate (ADC) against CD30 that is expressed in Hodgkin Reed–Sternberg cells and lymphoma tumor cells, has also shown a significant antitumor effect [75].

    Bispecific T-cell engagers

    Another fast-growing research for cancer targeted therapy is the development of bispecific T-cell engagers (BiTEs). BiTEs have a well-reported structure in which two single chain variable fragments bind to two different antigens, one for the immune cell receptor and the other for the cancer antigen, together by a peptide linker, in order to localize the immune response against cancerous cells [76]. The first FDA-approved BiTE for the treatment of ALL, is blinatumomab, which was approved in 2014 and has the ability to bind to anti-CD3 chain of T cells and anti-CD19 of B cells together [77]. The first trials that involved blinatumomab have gained unsuccessful outcomes, such as neurological complications and cytokine release syndrome [78]. Further studies have demonstrated that by reducing the prolonged doses at a level that fulfil both high cancer cell killing and tolerable level of toxicity can lead to effective and safe use of this drug against hematological malignancies [79,80].

    Recently, extended efforts have been made to engineer BiTEs targeting different multiple myeloma expressed antigens [81–84]. A study by Hipp et al. demonstrated a novel BiTE (BI 836909), whereby BCMAs, in other words, highly expressed antigens on multiple myeloma cells and malignant plasma cells but not on naive B cells, are connected to anti-CD3ε fragments of T cells [81]. Results showed that the upregulated expression of CD25 and CD69 indicated T cell activity in BCMA positive cells, whereas, the BCMA negative cells had no expression of such cytokines. In regard to cell lysis activity of BI 836909, BCMA-positive multiple myeloma cell lysis has increased substantially compared with the BCMA-negative cells. These results have shown the highly potent ability of BI 836909 to induce multiple myeloma cell lysis upon activation of T cells, while sparing the healthy B cells of which BCMA was not expressed [81].

    Immune checkpoint inhibitors

    Host immunosurveillance plays a crucial role in eradicating pathogens, as well as, reducing any autoimmune responses. Such processes are significantly mediated by the effector T cells, which requires a two-level activation or inhibition, to change their fate: costimulatory and coinhibitory signals. These signals can ensure a proper effector T-cell functioning, known as immune checkpoints [85]. For instance, activation of effector T cell requires binding to a peptide. It becomes fully activated once it binds to a ligand from the B7-1 family. Interestingly, PD-1 and CTLA4 receptors and their ligands constitute different groups in the CD28/B7-1 family [86].

    Host immunosurveillance can also identify cancer cells and eradicate them by inducing their apoptosis [87]. However, this process is often not impeccable. Different hematopoietic malignancies have been reported to upregulate the expression of the inhibitory molecules, PD-1 and CTLA4, thereby, escaping immunosurveillance [87]. Remarkably, targeting those immune checkpoint inhibitors have revolutionized cancer treatment. Earlier clinical trials of drugs targeting immune checkpoint inhibitors have been reviewed extensively [85–87]. Herein, we shed the light on recent clinical studies employing PD-1 inhibitors for the treatment of various hematopoietic malignancies.

    PD-1 inhibitors were used in many clinical trials as a potential treatment approach of different hematological malignancies, including multiple myeloma. Increased PD-1 signaling was found to be associated with multiple myeloma, by preventing immunosurveillance [88]. The PD-1 inhibitor, pembrolizumab, was recently used in a Phase IB clinical trial with R/R multiple myeloma (RRMM) [89]. Pembrolizumab is a humanized antibody that inhibits PD-1 receptor signaling by blocking its association with its ligands. Pembrolizumab is used as a monotherapy for RRMM and was shown to be generally safe with no significant severe adverse effects. However, 56% of patients (n = 17) exhibited stable disease with a maiden period of 4 months. Interestingly, all 17 patients were previously treated with the immunomodulator, lenalidomide and had at least one refractory disease. Thus, despite lacking the desired objective response rate, pembrolizumab might be a promising anti-RRMM therapy, especially when used in combination with other anticancer drugs.

    Classical Hodgkin lymphoma (cHL) is a hematopoietic malignancy characterized by amplified PD-L1 expression due to a genetic amplification of chromosome 9p24.1 [90]. The first line of treatment after the failure of the conventional therapy of R/R cHL is salvage chemotherapy in combination with autologous stem cell transplant (ASCT). The second line of treatment would be the use of brentuximab vedotin, which has a considerably high overall response rate (75%), but low durable response. Therefore, the effect of high PD-L1 and PD-L2 expression should be targeted by PD-1 antibodies.

    Recently, the effect of PD-1 blockade in R/R cHL patients was assessed in a Phase II clinical study [90]. The efficiency and safety of pembrolizumab were tested after ASCT in 30 patients suffering from R/R cHL. Interestingly, the study reported a PFS in 82% of patients accounting for 60–80% of PFS rates. Moreover, pembrolizumab has shown a considerably safe profile, with 40% of patients showing one grade 2 or higher immune-related adverse event, and 30% with one grade 3 adverse events.

    Nivolumab, an anti-PD-1 drug, was also used to assess its efficiency and safety on RR cHL patients [91]. Nivolumab combined with brentuximab vedotin before ASCT therapy has resulted in a considerably high objective response rate of 82% and an overall response rate of 61% (n = 61) [92]. Moreover, most reported adverse events are of grades 1 and 2, thus, indicating the safety of this treatment approach for R/R cHL.

    Primary mediastinal large B-cell lymphoma (PMBCL) is a subtype of large B-cell lymphoma characterized by an amplified PD-1 axis [90,91]. A study conducted by Armand et al. analyzed the results of Phase 1B and Phase II clinical trials to assess pembrolizumab's efficiency and safety profile on R/R PMBCL patients [93]. Interestingly, pembrolizumab was shown to be safe and resulted in a high response rate in R/R PMBCL patients. The reported objective response rate of the Phase 1B study was 48%, while it was 45% for the Phase II study. Moreover, the two trials demonstrated similar safety profiles of grade 3 or 4 drug-related adverse events in 24 and 23% of patients, respectively. Remarkably, the data from the two clinical studies have contributed and accelerated the FDA's approval of pembrolizumab as a drug for R/R PMBCL. All of which indicated the importance of targeting disease immune-biomarkers as a monotherapy or in combination with other treatment approaches.

    Immunomodulatory drugs

    Immunomodulatory drugs (IMiDs) are thalidomide analogues that combine higher immunomodulation and anticancer properties with less toxicity profiles [94]. Thalidomide was initially used for the treatment of multiple myeloma and has shown a significant anticancer activity via several mechanisms, including inhibition of angiogenesis and TNF-α synthesis [94]. Other IMiDs, such as lenalidomide and pomalidomide, are able to increase the production of IL-10, an anti-inflammatory cytokine, and to inhibit the production of pro-inflammatory cytokines such as TNF-α, IL-1β and IL-6. In fact, these analogues have been reported to be up to 50,000-fold more potent than thalidomide at inhibiting TNF-α production by peripheral blood mononuclear cells in vitro [95]. IMiDs can potently enhance the functions of T cells and NK cells by inducing IL-2 and interferon-γ production [96]. Particularly, lenalidomide has shown to increase the T-cell proliferation and cytokine production by enhancing AP-1 transcriptional activity [97], and reducing CTLA4 inhibition via directly inducing tyrosine phosphorylation of CD28 on T cells [98]. Other studies have also suggested that IMiDs can inhibit the activity of Treg cells [99], and can exert a CASP8 effect on multiple myeloma cells [100].

    Through the two past decades, many clinical trials have taken place to study the effect of IMiDs on several hematopoietic malignancies including multiple myeloma, acute myeloid leukemia (AML) and CLL (Table 1). A recent randomized, open-label Phase III study (Myeloma XI) included patients who are diagnosed newly with multiple myeloma, and have compared the PFS and overall survival of those who received induction therapy [101]. These patients were either assigned to maintenance therapy with lenalidomide or were kept under observation. The results demonstrated that the median PFS was significantly higher, while the overall survival was slightly higher for patients who received lenalidomide therapy. Both PFS and overall survival outcomes draw preferences to the use of lenalidomide as a maintenance therapy. However, 45% of patients receiving lenalidomide had high adverse effects, while only 17% of patients on observation had serious adverse effects [101]. Overall, the results of this study demonstrated the improvement of treatment outcomes using lenalidomide as a maintenance therapy.

    Table 1. Recent immunomodulatory drugs used in hematological malignancies that are complied with ClinicalTrials.gov (last accessed in July 2021).
    Drug(s)IndicationPhaseStageClinicalTrials.gov Identifier
    Lenalidomide, chlorambucilB-cell lymphocytic leukemia (n = 450)3Completed and last updated 9 July 2019NCT00910910
    DTRMWXHS-12, everolimus, pomalidomide• Relapsed CLL
    • Refractory CLL
    • Diffuse large B cell lymphoma
    • Follicular lymphoma (n = 120)
    2Recruiting and last updated 27 November 2020NCT04305444
    LenalidomideMultiple myeloma (n = 273)3Completed and last updated 11 May 2020NCT02322320
    Lenalidomide, dexamethasoneMultiple myeloma (n = 26)2Completed and last updated 8 November 2018NCT01698801
    LenalidomideAML (n = 17)2Completed and last updated 7 January 2020NCT02538965
    Pomalidomide, cytarabine, daunorubicin hydrochloride, etoposide, idarubicin hydrochloride• AML
    • Chronic myelomonocytic leukemia-2
    • High risk myelodysplastic syndrome
    • Myeloproliferative neoplasm (n = 50)
    1Completed and last updated 14 October 2020NCT02029950
    Pomalidomide (POMALYST®)Multiple myeloma (n = 600)Recruiting and last updated 11 May 2020NCT03288974

    AML: Acute myeloid leukemia; CLL: Chronic lymphocytic leukemia.

    In a Phase III trial, in which stem cell transplant-eligible patients who are newly diagnosed with multiple myeloma have received either lenalidomide with a low dose of dexamethasone until progression (LD), lenalidomide with a low dose of dexamethasone for 72 weeks (LD72), or melphalan with prednisone and thalidomide for 72 weeks (MPT). The findings of the 4-year PFS rate were considerably higher for LD group of patients compared with LD72 and MPT groups, which were comparable [102]. Moreover, LD and LD72 groups had higher 4-year overall survivals when compared with the MPT group [102].

    Lenalidomide was also tested intensively for the treatment of other hematologic malignancies. One study has reported the use of both lenalidomide and pomalidomide, both second-generation IMiDs, which have direct and immune-driven activities on AML. These activities were presented by the suppression of leukemia progress in vivo, AML cell death in vitro, and the increase of NK cell activity in slowing leukemia's development [103]. Furthermore, lenalidomide combined with other drugs was used to treat different lymphoma types. A Phase III study where rituximab was used with lenalidomide compared with a placebo, was shown an improved efficacy of rituximab in terms of PFS rate for patients with indolent non-Hodgkin's lymphoma [104].

    Another study featured the use of obinutuzumab along with lenalidomide in the treatment of R/R follicular B cell lymphoma. Out of 86 evaluable patients, 79% have reached a CR, which is the primary end point of the study, while 38% achieved an overall response by the end of the induction therapy [105]. Several studies have also reported the enhanced efficacy of using lenalidomide with different drugs in the treatment of CLL, in regards to altering the cytokines secretion [106], or reaching to higher complete responses at different doses within patients tolerable levels [107].

    Immunotherapeutic vaccines

    The concept of cancer therapeutic vaccination relies on the administration of TAAs, tumor specific antigens (TSAs) or whole cells to enhance active immunity of cancer patients through T-cells upregulation [108]. Despite the extensive clinical trials that took place in the 2000s, there is still no FDA approved therapeutic vaccines for the treatment of any type of hematological malignancies [109,110]. Nonetheless, more efforts have been conducted to develop a hematologic cancer vaccine by using different formulations. In general, therapeutic vaccines can be divided into cell-based or antigen specific vaccines, each of which holds a promising cancer targeting approach.

    Tumor specific vaccines aim at utilizing antigens, specifically expressed on the surface of cancer cells, in an attempt to allow immune cells to recognize and target tumor cells, while sparing healthy ones that do not express them. Cancer derived proteins and multi-peptides are the main constituents of this therapeutic approach, both of which can be stemmed from mutated, such as neoantigens, or non-mutated proteins [111]. Since the 1990's, more attention has been drawn toward the use of heat shock proteins (HSPs), which are overexpressed in many types of cancer cells that are associated with their proliferation and metastasis, in cancer vaccination [112]. HSP105, a therapeutic target for several cancer types, including non-Hodgkin lymphoma [113], has recently undergone Phase I clinical trial involving 30 patients diagnosed with either esophageal or colorectal cancers [114]. The level of HSP105 was recorded, and the results showed a clear declination of HSP105 level of expression for those who were vaccinated [114]. Other emerging studies have focused on using WT1 for AML patients [115,116]. A Phase II trial study of 22 AML patients with complete remission has reported that 47% of those patients reached an overall score of 3 years, and there were detectable CD4+ and CD8+ immunological responses of 44 and 86%, respectively. Thereby, WT1 protein was considered a potential AML targeting approach, while still calling for larger clinical trials to address other possible risks and success rates [115].

    Another highly investigated hematological cancer vaccine approach is the use of whole tumor cells to trigger the immune system rather than using specific components of the cells. This strategy of cancer immunization has the advantage in which the injection of whole cancer cells employs multiple cancer specific antigens as therapeutic targets all at once. However, the clear disadvantages of this approach are the difficulty of cell line harvesting and the high cost of the procedure [117]. A new study on AML mice model has described the use of Cryogel® (Aspen Aerogels, MA, USA) a microporous biomaterial, as a cancer vaccine. Cryogel was used as an immune cell activator and a delivery system for the combination of GM-CSF, TLR9 and AML-associated antigens, specifically WT1, which are the main immunogenic components [118]. The results proved the efficacy of the Cryogel in inducing an immunogenic response, with a full protection against AML challenge (day 0) and rechallenge (day 100), whereas there was no mice survival beyond 29 days. In addition, subcutaneous bolus injection of the immunogenic components (GM-CSF and antigen) showed lower number of CD11c+ and CD86+ dendritic cells in the draining lymph nodes compared with the Cryogel vaccine. The survival rate of the untreated group was lower (between 25 and 30 days), followed by the chemotherapy alone group (between 30 and 40 days), and finally, the chemotherapy with Cryogel group that showed 100% mice survival rate, despite the presence of the antigens indicating that this effect depends on the GM-CSF [118].

    Besides, the ability of dendritic cell-based vaccines to trigger both innate and adaptive immunities have favored their wide application on hematologic malignancies [119]. Several studies on the use of dendritic cell-based vaccines for AML patients have demonstrated high efficacy, in other words, triggering the immune system, and safety levels that might lead to their potential use as vaccines for hematopoietic malignancies [120,121].

    Conclusion & future perspective

    Blood cancer immunotherapeutic approaches have been a great success in recent years. This attributed to their distinguishable features, including their proximity to the immune cell microenvironment and the simplicity of their isolation and manipulation [122]. Several regimens of immunotherapy have been studied and approved by the FDA, while many are still in the late stages of clinical development, including adoptive cellular transfer, antibody-based therapies and immune checkpoint inhibitors. Many of these immunotherapies have been discussed in details as efficient therapeutic approaches for hematological malignancies.

    However, in spite of the enormous achievements of immunotherapy in hematological malignancies over the past years, managing their associated severe adverse effects is an unmet need to be solved and required further investigations. Combined the use of multiple immune therapeutics could offer a more effective therapeutic approach for hematological malignancies in the near future. Moreover, the precise identification of new molecular targets and new molecular pathways, to reduce the damage of normal cells, is still needed. Altogether, these future and supplemental studies will lead to the development of novel therapeutic approaches that will complement the existing treatment strategies and improve the overall longevity and lifestyle of cancer patients.

    Executive summary

    Immunotherapy against hematological malignancies

    • Immunotherapy has proven to improve the overall survival rate of cancer patients.

    • Over the past few years, this treatment approach against hematopoietic malignancies has shown massive clinical success over conventional approaches, despite being a newly growing field.

    • The clinical success of this approach attributes to the distinguishable features of immunotherapies such as their proximity to the immune cell microenvironment and the simplicity of their isolation and manipulation.

    • Many of these drugs and cell-based therapies are being approved by the US FDA and are in the late stages of clinical trials.

    Different types of immunotherapy

    • There are several immunotherapy approaches for hematological malignancy ranging from nonmyeloablative transplantation, T-cell immunotherapy, natural killer cells, and immune agonist to monoclonal antibodies and vaccination.

    • There is no ideal immunotherapeutic approach for the treatment of this type of cancers, as each approach has its own advantages and disadvantages.

    Combinational therapeutic approach

    • Despite of the vast successes of immunotherapeutic in hematological malignancies, managing the associated severe adverse effects of these agents is the major burden of this treatment approach.

    • Combining multiple immunotherapy agents might offer a more efficient therapeutic approach for this type of cancers.

    • This combinational approach could complement the existing therapeutic strategies and improve the overall longevity and lifestyle of cancer patients.

    Future perspective

    • There is a need to identify new precise molecular targets and pathways, in order to reduce the damage of normal cells, and thus, the associated side effects.

    • There is also a necessity of testing combined immunotherapies that might dramatically improve treatment outcomes in cancer patients.

    Financial & competing interests disclosure

    This work was supported by a grant from King Abdulaziz City for Science and Technology through the Center of Excellence for Biomedicine (CEBM). K Shah owns equity in and is a member of the Board of Directors of AMASA Therapeutics, a company developing stem cell-based therapies for cancer. K Shah's interests were reviewed and are managed by Brigham and Women's Hospital and Partners HealthCare in accordance with their conflict of interest policies. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Jagannathan-Bogdan M, Zon LI. Hematopoiesis. Development 140(12), 2463–2467 (2013).
    • 2. Doulatov S, Notta F, Laurenti E, Dick JE. Hematopoiesis: a human perspective. Cell Stem Cell 10(2), 120–136 (2012).
    • 3. Jurlander J. Hematological malignancies, leukemias and lymphomas. In: Encyclopedia of Cancer. Schwab M (Ed.)., Springer-Verlag, Berlin/Heidelberg, Germany, 1640–1644 (2011).
    • 4. Hu D, Shilatifard A. Epigenetics of hematopoiesis and hematological malignancies. Genes Development 30(18), 2021–2041 (2016).
    • 5. Zaidan N, Ottersbach K. The multi-faceted role of Gata3 in developmental haematopoiesis. Open Biol. 8(11), 180152 (2018).
    • 6. Lawrence MS, Stojanov P, Mermel CH et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505(7484), 495–501 (2014).
    • 7. Cai SF, Chen C-W, Armstrong SA. Drugging chromatin in cancer: recent advances and novel approaches. Mol. Cell 60(4), 561–570 (2015).
    • 8. Li Z-W, Dalton WS. Tumor microenvironment and drug resistance in hematologic malignancies. Blood Rev. 20(6), 333–342 (2006).
    • 9. Raffaghello L, Vacca A, Pistoia V, Ribatti D. Cancer associated fibroblasts in hematological malignancies. Oncotarget 6(5), 2589–2603 (2015).
    • 10. Cheng H, Sun G, Cheng T. Hematopoiesis and microenvironment in hematological malignancies. Cell Regen. 7(1), 22–26 (2018).
    • 11. Kumar B, Garcia M, Murakami JL, Chen C-C. Exosome-mediated microenvironment dysregulation in leukemia. Biochim. Biophys. Acta 1863(3), 464–470 (2016).
    • 12. Sánchez-Aguilera A, Méndez-Ferrer S. The hematopoietic stem-cell niche in health and leukemia. Cell. Mol. Life Sci. 74(4), 579–590 (2017).
    • 13. Glait-Santar C, Desmond R, Feng X et al. Functional niche competition between normal hematopoietic stem and progenitor cells and myeloid leukemia cells. Stem Cells 33(12), 3635–3642 (2015).
    • 14. Miraki-Moud F, Anjos-Afonso F, Hodby KA et al. Acute myeloid leukemia does not deplete normal hematopoietic stem cells but induces cytopenias by impeding their differentiation. Proc. Natl Acad. Sci. USA 110(33), 13576–13581 (2013).
    • 15. Colmone A, Amorim M, Pontier AL, Wang S, Jablonski E, Sipkins DA. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science 322(5909), 1861–1865 (2008).
    • 16. Atilla E, Ataca Atilla P, Demirer T. A Review of myeloablative vs reduced intensity/non-myeloablative regimens in allogeneic hematopoietic stem cell transplantations. Balkan Med. J. 34(1), 1–9 (2017). •• This reference shares unambiguous information focused on recent studies and clinical trials in the field, through exploring wider range of regiment approaches followed by intensive review of clinical outcomes.
    • 17. Nagler A, Shimoni A. Conditioning. In: The EBMT Handbook: Hematopoietic Stem Cell Transplantation and Cellular Therapies (7th Edition). Carreras EDufour CMohty MKröger N (Eds). Springer International Publishing, Cham, Switzerland, 99–107 (2019).
    • 18. Luger SM, Ringdén O, Zhang M-J et al. Similar outcomes using myeloablative vs reduced-intensity allogeneic transplant preparative regimens for AML or MDS. Bone Marrow Transplant. 47(2), 203–211 (2012).
    • 19. Bacigalupo A, Ballen K, Rizzo D et al. Defining the intensity of conditioning regimens: working definitions. Biol. Blood Marrow Transplant. 15(12), 1628–1633 (2009).
    • 20. Storb R, Sandmaier BM. Nonmyeloablative allogeneic hematopoietic cell transplantation. Haematologica 101(5), 521–530 (2016).
    • 21. Saraf SL, Oh AL, Patel PR et al. Nonmyeloablative stem cell transplantation with alemtuzumab/low-dose irradiation to cure and improve the quality of life of adults with sickle cell disease. Biol. Blood Marrow Transplantation. 22(3), 441–448 (2016).
    • 22. Guilcher GMT, Truong TH, Saraf SL, Joseph JJ, Rondelli D, Hsieh MM. Curative therapies: allogeneic hematopoietic cell transplantation from matched related donors using myeloablative, reduced intensity, and nonmyeloablative conditioning in sickle cell disease. Semin. Hematol. 55(2), 87–93 (2018).
    • 23. Vaughn JE, Sorror ML, Storer BE et al. Long-term sustained disease control in patients with mantle cell lymphoma with or without active disease after treatment with allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. Cancer 121(20), 3709–3716 (2015).
    • 24. Ahmad I, Leblanc R, Cohen S et al. Favorable long-term outcome of patients with multiple myeloma using a frontline tandem approach with autologous and non-myeloablative allogeneic transplantation. Bone Marrow Transplant. 51(4), 529–535 (2016).
    • 25. Puronen CE, Cassaday RD, Stevenson PA et al. Long-term follow-up of 90y-ibritumomab tiuxetan, fludarabine, and total body irradiation-based nonmyeloablative allogeneic transplant conditioning for persistent high-risk b cell lymphoma. Biol. Blood Marrow Transplant. 24(11), 2211–2215 (2018).
    • 26. Storb R, Sandmaier BM. Nonmyeloablative allogeneic hematopoietic cell transplantation. Haematologica 101(5), 521–530 (2016).
    • 27. Storb R. HSCT: historical perspective. In: The EBMT Handbook: Hematopoietic Stem Cell Transplantation and Cellular Therapies (7th Edition). Carreras EDufour CMohty MKröger N (Eds). Springer International Publishing, Cham, Switzerland, 3–9 (2019).
    • 28. Storb RF, Champlin R, Riddell SR, Murata M, Bryant S, Warren EH. Non-myeloablative transplants for malignant disease. Hematology Am. Soc. Hematol. Educ. Program 2001(1), 375–391 (2001).
    • 29. Cheadle EJ, Gornall H, Baldan V, Hanson V, Hawkins RE, Gilham DE. CAR T cells: driving the road from the laboratory to the clinic. Immunol. Rev. 257(1), 91–106 (2014).
    • 30. Kumar BV, Connors TJ, Farber DL. Human T cell development, localization, and function throughout life. Immunity 48(2), 202–213 (2018).
    • 31. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 20(11), 651–668 (2020).
    • 32. Aleksic M, Liddy N, Molloy PE et al. Different affinity windows for virus and cancer-specific T-cell receptors: implications for therapeutic strategies. Eur. J. Immunol. 42(12), 3174–3179 (2012).
    • 33. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene 27(45), 5904–5912 (2008).
    • 34. Wang M, Yin B, Wang HY, Wang RF. Current advances in T-cell-based cancer immunotherapy. Immunotherapy 6(12), 1265–1278 (2014).
    • 35. Borrello I, Noonan KA. Marrow-infiltrating lymphocytes – Role in biology and cancer therapy. Front. Immunol. 7(MAR), 112 (2016).
    • 36. Feuerer M, Beckhove P, Garbi N et al. Bone marrow as a priming site for T-cell responses to blood-borne antigen. Nat. Med. 9(9), 1151–1157 (2003).
    • 37. Noonan KA, Huff CA, Davis J et al. Adoptive transfer of activated marrow-infiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma. Sci. Trans. Med. 7(288), 288ra78–288ra78 (2015). •• This is the first report of the clinical use of marrow-infiltrating lymphocytes adoptive therapy, demonstrating its efficacy and feasibility in the treatment of multiple myeloma.
    • 38. Noonan K, Matsui W, Serafini P et al. Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors. Cancer Res. 65(5), 2026–2034 (2005).
    • 39. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17(3), 147–167 (2020). • Explains innovative chimeric antigen receptor (CAR) engineering strategies such as engineering multi-specific CAR T cells targeting to circumvent antigen loss or heterogeneity in hematological malignancies.
    • 40. Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am. J. Hematol. 94(S1), S3–S9 (2019).
    • 41. Garrido F, Aptsiauri N, Doorduijn EM, Garcia Lora AM, van Hall T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr. Opin. Immunol. 39, 44–51 (2016).
    • 42. Rohaan MW, Wilgenhof S, Haanen JBAG. Adoptive cellular therapies: the current landscape. Virchows Archiv. 474(4), 449–461 (2019).
    • 43. Holstein SA, Lunning MA. CAR T-cell therapy in hematologic malignancies: a voyage in progress. Clin. Pharmacol. Ther. 107(1), 112–122 (2020).
    • 44. Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat. Rev. Clin. Oncol. 13(5), 273–290 (2016). • Extensive review of latest immunotherapeutic approaches for treating cancer including a summary for the clinical trials using CD19 CAR T-Cell therapy for B cell acute lymphoblastic leukemia patients.
    • 45. Ghorashian S, Pule M, Amrolia P. CD19 chimeric antigen receptor T cell therapy for haematological malignancies. Br. J. Haematol. 169(4), 463–478 (2015).
    • 46. 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 116(20), 4099–4102 (2010).
    • 47. Kochenderfer JN, Dudley ME, Kassim SH et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J. Clin.l Oncol. 33(6), 540–549 (2015). •• This is the first report of successful treatment of diffuse large B cell lymphoma using anti-CD19 CAR T-cell therapy, with eight out of 15 patients achieving complete remission.
    • 48. Brentjens RJ, Davila ML, Riviere I et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Trans. Med. 5(177), 177ra38 (2013).
    • 49. Davila ML, Riviere I, Wang X et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Trans. Med. 6(224), 224ra25 (2014).
    • 50. Porter DL, Hwang WT, Frey NV et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Trans. Med. 7(303), 303ra139 (2015).
    • 51. Zhang Q, Ping J, Huang Z et al. CAR-T cell therapy in cancer: tribulations and road ahead. J. Immunol. Res. 2020, 1924379 (2020).
    • 52. Kalaitsidou M, Kueberuwa G, Schütt A, Gilham DE. CAR T-cell therapy: toxicity and the relevance of preclinical models. Immunotherapy 7(5), 487–497 (2015).
    • 53. Bailey SR, Maus MV. Gene editing for immune cell therapies. Nat. Biotechnol. 37(12), 1425–1434 (2019).
    • 54. Wang N, Hu X, Cao W et al. Efficacy and safety of CAR19/22 T-cell cocktail therapy in patients with refractory/relapsed B-cell malignancies. Blood 135(1), 17–27 (2020).
    • 55. Dai H, Wu Z, Jia H et al. Bispecific CAR-T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J. Hematol. Oncol. 13(1), 30 (2020).
    • 56. Vivier E, Raulet DH, Moretta A et al. Innate or adaptive immunity? The example of natural killer cells. Science 331(6013), 44–49 (2011).
    • 57. Freud AG, Mundy-Bosse BL, Yu J, Caligiuri MA. The broad spectrum of human natural killer cell diversity. Immunity 47(5), 820–833 (2017).
    • 58. Mariotti FR, Quatrini L, Munari E, Vacca P, Moretta L. Innate lymphoid cells: expression of PD-1 and other checkpoints in normal and pathological conditions. Front. Immunol. 10, 910 (2019).
    • 59. Cerwenka A, Lanier LL. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol. 16(2), 112–123 (2016).
    • 60. Orange JS. Formation and function of the lytic NK-cell immunological synapse. Nat. Rev. Immunol. 8(9), 713–725 (2008).
    • 61. Sivori S, Meazza R, Quintarelli C et al. NK cell-based immunotherapy for hematological malignancies. J. Clin. Med. 8(10), 1702 (2019).
    • 62. Bernardini G, Antonangeli F, Bonanni V, Santoni A. Dysregulation of chemokine/chemokine receptor axes and nk cell tissue localization during diseases. Front. Immunol. 7, 402 (2016).
    • 63. Kärre K. NK Cells, MHC class I molecules and the missing self. Scand. J. Immunol. 55(3), 221–228 (2002).
    • 64. Myers JA, Miller JS. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Immunol. 18(2), 85–100 (2020).
    • 65. Daher M, Rezvani K. Next generation natural killer cells for cancer immunotherapy: the promise of genetic engineering. Curr. Opin. Immunol. 51, 146–153 (2018).
    • 66. Tonn T, Schwabe D, Klingemann HG et al. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy 15(12), 1563–1570 (2013).
    • 67. Herrera L, Santos S, Vesga MA et al. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Sci. Rep. 9(1), 18729 (2019).
    • 68. Xie G, Dong H, Liang Y, Ham JD, Rizwan R, Chen J. CAR-NK cells: a promising cellular immunotherapy for cancer. EBioMedicine 59, 102975 (2020).
    • 69. Liu E, Marin D, Banerjee P et al. Use of CAR-transduced natural killer cells inCD19-positive lymphoid tumors. N. Engl. J. Med. 382(6), 545–553 (2020).
    • 70. Shimasaki N, Jain A, Campana D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 19(3), 200–218 (2020).
    • 71. Pierpont TM, Limper CB, Richards KL. Past, present, and future of rituximab – the world's first oncology monoclonal antibody therapy. Front. Oncol. 8, 163 (2018).
    • 72. Wierda WG, Kipps TJ, Mayer J et al. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J. Clin. Oncol. 28(10), 1749–1755 (2010).
    • 73. Goede V, Fischer K, Busch R et al. Obinutuzumab plus chlorambucil in patients with cll and coexisting conditions. N. Engl. J. Med. 370(12), 1101–1110 (2014).
    • 74. de Weers M, Tai Y-T, van der Veer MS et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J. Immunol. 186(3), 1840–1848 (2011).
    • 75. Younes A, Bartlett NL, Leonard JP et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N. Engl. J. Med. 363(19), 1812–1821 (2010).
    • 76. Mack M, Riethmuller G, Kufer P. A small bispecific antibody construct expressed as a functional single-chain molecule with high tumor cell cytotoxicity. Proc. Natl Acad. Sci. USA 92(15), 7021–7025 (1995).
    • 77. Zhu M, Wu B, Brandl C et al. Blinatumomab, a Bispecific T-cell Engager (BiTE®) for CD-19 targeted cancer immunotherapy: clinical pharmacology and its implications. Clin. Pharmacokinet. 55(10), 1271–1288 (2016).
    • 78. Oak E, Bartlett NL. Blinatumomab for the treatment of B-cell lymphoma. Expert Opin. Investig. Drugs 24(5), 715–724 (2015).
    • 79. Bargou R, Leo E, Zugmaier G et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321(5891), 974–977 (2008).
    • 80. Goebeler M-E, Knop S, Viardot A et al. Bispecific T-Cell Engager (BiTE) antibody construct blinatumomab for the treatment of patients with relapsed/refractory non-hodgkin lymphoma: final results from a Phase I study. J. Clin. Oncol. 34(10), 1104–1111 (2016).
    • 81. Hipp S, Tai Y-T, Blanset D et al. A novel BCMA/CD3 bispecific T-cell engager for the treatment of multiple myeloma induces selective lysis in vitro and in vivo. Leukemia 31(8), 1743–1751 (2016).
    • 82. Hipp S, Deegen P, Wahl J et al. BI 836909, a novel bispecific T cell engager for the treatment of multiple myeloma induces highly specific and efficacious lysis of multiple myeloma cells in vitro and shows anti-tumor activity in vivo. Blood 126(23), 2999–2999 (2015).
    • 83. Richter JR, Landgren CO, Kauh JS et al. Phase 1, multicenter, open-label study of single-agent bispecific antibody T-cell engager GBR 1342 in relapsed/refractory multiple myeloma. J. Clin. Oncol. 36(Suppl. 15), TPS3132–TPS3132 (2018).
    • 84. Topp M, Duell J, Zugmaier G et al. S825 evaluation of AMG 420, an Anti-BCMA Bispecific T-cell Engager (BITE®) immunotherapy, in R/R multiple myeloma (MM) patients: updated results of a first-in-human (FIH) Phase 1 dose escalation study. HemaSphere 3(S1), 365 (2019).
    • 85. Pianko MJ, Goldberg AD, Lesokhin AM. Clinical development of PD-1 blockade in hematologic malignancies. Cancer 24(1), 31–35 (2018).
    • 86. Wang H, Kaur G, Sankin AI, Chen F, Guan F, Zang X. Immune checkpoint blockade and CAR-T cell therapy in hematologic malignancies. J. Hematol. Oncol. 12(1), 59 (2019).
    • 87. Ok CY, Young KH. Checkpoint inhibitors in hematological malignancies. J. Hematol. Oncol. 10(1), 103 (2017).
    • 88. Neri P, Bahlis NJ, Lonial S. New Strategies in multiple myeloma: immunotherapy as a novel approach to treat patients with multiple myeloma. Clin. Cancer Res. 22(24), 5959–5965 (2016).
    • 89. Ribrag V, Avigan DE, Green DJ et al. Phase 1b trial of pembrolizumab monotherapy for relapsed/refractory multiple myeloma: KEYNOTE-013. Br. J. Haematol. 186(3), e41–e44 (2019).
    • 90. Armand P, Chen Y-B, Redd RA et al. PD-1 blockade with pembrolizumab for classical Hodgkin lymphoma after autologous stem cell transplantation. Blood 134(1), 22–29 (2019). •• A clinical trial has met their primary end point with a high response rate and low toxicity profile.
    • 91. Ansell SM, Minnema MC, Johnson P et al. Nivolumab for relapsed/refractory diffuse large B-cell lymphoma in patients ineligible for or having failed autologous transplantation: a single-arm, Phase II study. J. Clin. Oncol. 37(6), 481–489 (2019).
    • 92. Herrera AF, Moskowitz AJ, Bartlett NL et al. Interim results of brentuximab vedotin in combination with nivolumab in patients with relapsed or refractory Hodgkin lymphoma. Blood 131(11), 1183–1194 (2018). •• A clinical trial has met their primary end point with a high response rate and low toxicity profile.
    • 93. Armand P, Rodig S, Melnichenko V et al. Pembrolizumab in relapsed or refractory primary mediastinal large B-cell lymphoma. J. Clin. Oncol. 37(34), 3291–3299 (2019).
    • 94. Zangari M, Elice F, Tricot G. Immunomodulatory drugs in multiple myeloma. Expert Opin. Investig. Drugs 14(11), 1411–1418 (2005).
    • 95. Corral LG, Haslett PA, Muller GW et al. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-alpha. J. Immunol. 163(1), 380–386 (1999).
    • 96. Hayashi T, Hideshima T, Akiyama M et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. Br. J. Haematol. 128(2), 192–203 (2005).
    • 97. Schafer PH, Gandhi AK, Loveland MA et al. Enhancement of cytokine production and AP-1 transcriptional activity in T cells by thalidomide-related immunomodulatory drugs. J. Pharmacol. Exp. Ther. 305(3), 1222–1232 (2003).
    • 98. LeBlanc R, Hideshima T, Catley LP et al. Immunomodulatory drug costimulates T cells via the B7-CD28 pathway. Blood 103(5), 1787–1790 (2004).
    • 99. Galustian C, Meyer B, Labarthe M-C et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol. Immunother. 58(7), 1033–1045 (2009).
    • 100. Mitsiades N, Mitsiades CS, Poulaki V et al. Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications. Blood 99(12), 4525–4530 (2002).
    • 101. Jackson GH, Davies FE, Pawlyn C et al. Lenalidomide maintenance versus observation for patients with newly diagnosed multiple myeloma (Myeloma XI): a multicentre, open-label, randomised, Phase III trial. Lancet Oncol. 20(1), 57–73 (2019). •• A recent and largest clinical trial using lenalidomide drug as maintenance therapy for multiple myeloma patients.
    • 102. Facon T, Dimopoulos MA, Dispenzieri A, Catalano J, Belch AR, Cavo M. Final survival analysis from the first trial: lenalidomide plus low-dose dexamethasone until progression (rd cont) v melphalan, prednisone and thalidomide (mpt), and rd for 18 cycles (rd18) for transplant-ineligible (tne) patients (pts) with newly diagnosed multiple myeloma. Clin. Lymphoma Myeloma Leuk. 17(1), e63–e64 (2017).
    • 103. Le Roy A, Prébet T, Castellano R et al. Immunomodulatory drugs exert anti-leukemia effects in acute myeloid leukemia by direct and immunostimulatory activities. Front. Immunol. 9, 977 (2018).
    • 104. Leonard JP, Trneny M, Izutsu K et al. AUGMENT: a Phase III study of lenalidomide plus rituximab versus placebo plus rituximab in relapsed or refractory indolent lymphoma. J. Clin. Oncol. 37(14), 1188–1199 (2019).
    • 105. Morschhauser F, Le Gouill S, Feugier P et al. Obinutuzumab combined with lenalidomide for relapsed or refractory follicular B-cell lymphoma (GALEN): a multicentre, single-arm, Phase II study. Lancet Haematol. 6(8), e429–e437 (2019).
    • 106. Chen CI, Paul H, Snitzler S et al. A Phase II study of lenalidomide and dexamethasone in previously untreated patients with chronic lymphocytic leukemia (CLL). Leuk. Lymphoma 60(4), 980–989 (2018).
    • 107. Soumerai JD, Davids MS, Werner L et al. Phase I study of lenalidomide, bendamustine, and rituximab in previously untreated patients with chronic lymphocytic leukemia. Leuk. Lymphoma 60(12), 2931–2938 (2019).
    • 108. DeMaria PJ, Bilusic M. Cancer Vaccines. Hematol. Oncol. Clin. North Am. 33(2), 199–214 (2019).
    • 109. Trudel S, Li Z, Dodgson C et al. Adenovector engineered interleukin-2 expressing autologous plasma cell vaccination after high-dose chemotherapy for multiple myeloma – a Phase 1 study. Leukemia 15(5), 846–854 (2001).
    • 110. Younes A. A Phase II study of heat shock protein–peptidecomplex–96 vaccine therapy in patients with indolent non-hodgkin's lymphoma. Clin. Lymphoma 4(3), 183–185 (2003).
    • 111. Engelhard VH, Obeng RC, Cummings KL et al. MHC-restricted phosphopeptide antigens: preclinical validation and first-in-humans clinical trial in participants with high-risk melanoma. J. Immunother. Cancer 8(1), e000262 (2020).
    • 112. Srivastava PK, Udono H. Heat shock protein-peptide complexes in cancer immunotherapy. Curr. Opin. Immunol. 6(5), 728–732 (1994).
    • 113. Zappasodi R, Ghedini GC, Bongarzone I et al. Serological identification of HSP105 as a novel non-Hodgkin lymphoma therapeutic target. Blood 116(21), 463–463 (2010).
    • 114. Shimizu Y, Yoshikawa T, Kojima T et al. Heat shock protein 105 peptide vaccine could induce antitumor immune reactions in a Phase I clinical trial. Cancer Sci. 110(10), 3049–3060 (2019).
    • 115. Maslak PG, Dao T, Bernal Y et al. Phase II trial of a multivalent WT1 peptide vaccine (galinpepimut-S) in acute myeloid leukemia. Blood Adv. 2(3), 224–234 (2018).
    • 116. Tsuboi A, Oka Y, Kyo T et al. Long-term WT1 peptide vaccination for patients with acute myeloid leukemia with minimal residual disease. Leukemia 26(6), 1410–1413 (2011).
    • 117. Singh M, Salnikova M. Novel Approaches and Strategies for Biologics, Vaccines and Cancer Therapies. Elsevier Inc, Amsterdam, The Netherlands (2015).
    • 118. Shah NJ, Najibi AJ, Shih T-Y et al. A biomaterial-based vaccine eliciting durable tumour-specific responses against acute myeloid leukaemia. Nat. Biomed. Eng. 4(1), 40–51 (2020).
    • 119. Saxena M, Bhardwaj N. Re-emergence of dendritic cell vaccines for cancer treatment. Trends Cancer 4(2), 119–137 (2018).
    • 120. van de Loosdrecht AA, van Wetering S, Santegoets SJAM et al. A novel allogeneic off-the-shelf dendritic cell vaccine for post-remission treatment of elderly patients with acute myeloid leukemia. Cancer Immunol. Immunother. 67(10), 1505–1518 (2018).
    • 121. Hsu JL, Bryant CE, Papadimitrious MS et al. A blood dendritic cell vaccine for acute myeloid leukemia expands anti-tumor T cell responses at remission. Oncoimmunology 7(4), e1419114 (2018).
    • 122. Bachireddy P, Burkhardt UE, Rajasagi M, Wu CJ. Haematological malignancies: at the forefront of immunotherapeutic innovation. Nat. Rev. Cancer 15(4), 201–215 (2015).