Skip to main content

Interferon-alpha-based immunotherapies in the treatment of B cell-derived hematologic neoplasms in today’s treat-to-target era

Abstract

B cell lymphoma and multiple myeloma (MM) are the most common hematological malignancies which benefit from therapeutic monoclonal antibodies (mAbs)-based immunotherapies. Despite significant improvement on patient outcome following the use of novel therapies for the past decades, curative treatment is unavailable for the majority of patients. For example, the 5-year survival of MM is currently less than 50%. In the 1980s, interferon-α was used as monotherapy in newly diagnosed or previously treated MM with an overall response rate of 15–20%. Noticeably, a small subset of patients who responded to long-term interferon-α further achieved sustained complete remission. Since 1990, interferon-α-containing regimens have been used as a central maintenance strategy for patients with MM. However, the systemic administration of interferon-α was ultimately limited by its pronounced toxicity. To address this, the selective mAb-mediated delivery of interferon-α has been developed to enhance specific killing of MM and B-cell malignant cells. As such, targeted interferon-α therapy may improve therapeutic window and sustain responses, while further overcoming suppressive microenvironment. This review aims to reinforce the role of interferon-α by consolidating our current understanding of targeting interferon-α with tumor-specific mAbs for B cell lymphoma and myeloma.

Background

B-cell neoplasms account for about 80% of lymphomas, which are the most common type of blood cancer. In late 1990s, a new era of monoclonal antibody (mAb)-based immunotherapy emerged with the first anti-CD20 mAb for treatment of B-cell lymphomas. Surface CD20, a pan B-cell marker, is expressed during most stages of B cell development: on late pro-B cells to naïve, mature, and memory B cells; but not on precursor B cells, early pro-B cells, plasma blasts or plasma cells. Accordingly, anti-CD20 mAbs directly deplete B cells of intermediate stages whilst sparing pre-B cells and long-lived plasma cells, which highly expressed cell-surface CD38 instead. Multiple myeloma (MM), the second most common blood cancer, is a distinct B-cell derived neoplasm characterized by expansion of plasma cells in bone marrow. The mAb therapies have become available for MM patients by targeting SLAM Family Member 7 (SLAMF7) [1, 2] and CD38 [3], both of which highly express on primary MM cells. Specifically, anti-CD38 mAb daratumumab is the first mAb showing activity as a monotherapy in MM [3]. A very recent interim analysis of the phase 3 CASTOR trial also showed that therapeutic anti-CD38 mAb, when combined with bortezomib and dexamethasone, can significantly prolong progressive-free survival (PFS) in patients with early relapsed and/or refractory MM [4]. Although these mAb-based immunotherapies have led to significant improvements in treatment of B-cell neoplasms, patients may relapse and ultimately succumb to the disease. Therefore, the timely identification of an alternative approach, which more effectively destroys tumor cells including cancer stem cells (CSCs), is urgently needed.

Significant research efforts spanning nearly 4 decades have explored usage of cytokines as immunomodulators to enhance host immune response against cancer. The most commonly used cytokines are the interferons (IFN), named in 1957 based on their ability to “interfere” with viral replication in infected cells [5]. IFNs were then further classified into 3 pleiotropic polypeptides: type I, II, and III (Table 1) [5, 6]. In 1978, type I IFNs from supernatant of human leukocytes exposed to viruses were purified to homogeneity and sequenced, leading to the discovery of various subtypes of IFNs [7]. In 1981, recombinant IFNs were successfully expressed by Genentech, allowing for the large scale production of “clean” IFNs to meet both research and clinical demands [8, 9]. Interferon-alpha (IFN-α), a type I IFN, is the first recombinant subtype and also the most commonly used IFN in anti-cancer therapy. IFN-α comprises a family of more than 20 related but distinct members encoded by a cluster on chromosome 9. Among these, the most frequently used is IFN-α2, having 3 recombinant variants (α2a, α2b, α2c) depending upon the cells of origin [10]. IFN-α2b is the predominant variant in human genome.

Table 1 Subtypes of IFN family

IFN-α can be secreted by intratumoural dendritic cells (DCs) and malignant cells in response to various stimuli and via positive autocrine and paracrine loops. As reported, a majority of human B-lineage cell lines (e.g. lymphoblastoid cells, B lymphoma, and MM cells) spontaneously produce significant amounts of IFN-α [11]. Plasmacytoid DCs have earned the moniker “human IFN-producing cells” (IPCs), hence they have the greatest capacity to secrete type I IFNs. In the classical model of an antiviral immune response, IPCs are involved at two stages: (1) during the initial innate immune response stage, IPCs rapidly secrete type I IFNs to promote the function of natural killer (NK) cells, B cells, T cells, and myeloid DCs, and (2) at a later stage involving the adaptive immune response, IPCs differentiate into mature DC, which in turn directly regulates the function of T cells. All known IFN-α subtypes exert their function through a specific cell surface membrane receptor complex known as IFN-α receptor (IFN-ΑR), commonly designated as IFN α/β receptor. IFN-ΑRs consist of two high affinity chains: a 110 kDa subunit α (IFN-ΑR1) reported in 1990; and a 102 kDa subunit β (IFN-ΑR2c) reported in 1994. Additionally, two different spliced isoforms of IFN-ΑR subunit β have been reported: (1) 40 kDa soluble IFN-ΑR2a; and (2) 5 kDa transmembrane short form, IFN-ΑR2b. IFN-α binding to IFN-ΑRs leads to activation of intracellular signaling cascades that increase the expression and promote the activation of signal transducers and activators of transcription (STAT)1, STAT2, and STAT3. STAT1 is required for IFN-α-mediated cell death. IFN-ΑRs are expressed not only on malignant cells but also on non-malignant cells, which contributes to anti-tumor effects and nonspecific toxicity by IFN-α.

Systemic IFN-α administration is, to a large extent, hampered by its short half-life, high myelotoxicity, and paradoxical immunosuppressive effects. At present, cell-based immunotherapy is a very promising therapeutic approach; incorporation of a cell-based approach that exploits the specificity of mAb-targeting can selectively deliver IFN-α into the tumor compartment, with fewer side effects as normal cells are spared. This review thereby reevaluates the utility of IFN-α-based regimens for B-cell lymphoma and MM in the current treat-to-target era.

Preclinical studies of IFN-α in B cell lymphoma and myeloma

Recombinant IFN-α has shown activity against B-cell hematologic neoplasms, primarily through indirect depletion of B-cell neoplasms by immune activation of IFN-ΑR-expressing immune effector cells [12]. For T cells, IFN-α induces the generation and long-term survival of both cytotoxic CD8+ T cell (CTL) and memory CD8+ T cells against tumor antigens, as well as polarizes immune responses towards CD4+ T helper-1 (Th1) phenotype. For NK cells, IFN-α enhances NK cell-mediated toxicity and survival of NK cells. For B cells, IFN-α positively regulates antibody production. For DC cells, IFN-α promotes their maturation and chemotaxis. Moreover, IFN-α treatment induces the expression of programmed cell death-1 (PD-1) on tumor-infiltrating T cells and PD-L1 on tumors [13], which can be neutralized using checkpoint blockade with anti-PD-1/PD-L1 mAbs [14, 15], currently in clinical trials in both lymphoma and MM.

IFN-α can also trigger direct anti-tumor cytotoxicity. By activating IFN-ΑR signaling in B cell lymphomas, IFN-α can induce apoptosis [16], inhibit proliferation [17] and cell cycle progression [18], and promote terminal differentiation in cancer cells [19]. IFN-α signaling also upregulates major histocompatibility complex class I molecules on the surface of tumor cells, leading to enhanced tumor recognition by CTLs. IFN-α was recently reported to upregulate the expression of tumor-associated antigens on human breast cancer xenografts, highlighting their potential for synergy with mAb therapy [20].

However, the precise mechanisms underlying IFN-α’s anti-myeloma effect remain unclear [21]. This is due, in part, to contradictory reports on the effects of IFN-α on ex vivo cultured myeloma cells: some studies showed that IFN-α induces apoptosis and inhibits growth on myeloma cell lines [22], while other studies reported that IFN-α is a survival factor for human myeloma cells via upregulation of anti-apoptotic molecule Mcl-1 [23]. There is an ongoing debate questioning relevance of an ex vivo system to model the highly complex tumor microenvironment in MM [24]. The utility of IFN-α as a maintenance drug for patients with MM was first reported in 1990 [25]. Since then, multiple studies to define the therapeutic benefit of IFN-α-based maintenance regimens have had conflicting results. The primary focus of maintenance therapy in MM is to improve PFS and overall survival (OS). The achievement of positive responses hinges on the thorough depletion of CSC pools (i.e. myeloma-initiating cells) after minimal residual disease (MRD) is achieved following induction therapy. Residual myeloma cells can survive by senescence and entering into the quiescent G0 phase of the cell cycle [26], while IFN-α initially induced cell cycle arrest at the G0/G1 phase in an in vivo mouse model. Thereby, chronic stimulation by IFN-α could cause dormant hematopoietic stem cells to efficiently exit G0, and reverse therapeutic-induced senescence and drug-resistance [27]. IFN-α can therefore exert direct anti-cancer effects by re-activating and mobilizing senescent CSCs [28], which may explain the efficacy of IFN-α maintenance therapy in MM.

History of IFN-α-based therapy in B cell lymphoma and myeloma

In the field of B cell lymphoma and MM, the usage of IFN-α-based immunotherapies spans two distinct eras: (1) pre-mAb; and (2) post-mAb eras (Table 2). The utilization of IFN-α in the treatment of human B cell lymphoma dates back to the late 1970s, beginning with the use of natural IFN-α in murine models of leukemia and lymphoma (Additional file 1: Table S1) [2931]. In 1981, the National Cancer Institute undertook Phase II trials of IFN-α2a in patients with low-grade non-Hodgkin’s lymphoma (NHL) [32]. And then, IFN-α has been used mainly in the treatment of low-grade follicular lymphoma (FL), the most common indolent NHL. The efficacy of IFN-α in cutaneous T cell lymphoma (CTCL) was first reported in 1984 and also subsequently at the 1995 International Conference on CTCL to be the most effective single agent treatment. However, initial results of IFN-α treatment of other B-cell neoplasms were far less impressive, since IFN could provide only palliative benefit in certain low-grade or early stage B-cell lymphomas, with complete remission and overall response of 10 and 48%, respectively [33, 35]. The first reported instance of IFN-α use in human MM dates back to 1979 when Mellstedt et al. demonstrated its efficacy in previously untreated myeloma [36]. Since then, IFN-α2b has achieved 50 and 15% responses in patients with newly diagnosed and refractory MM, respectively [37]. From 1997 onwards, the introduction of anti-CD20 mAbs led to significantly better disease control in high-grade lymphoma subtypes (e.g. diffuse large B cell lymphoma) and advanced stage/high-grade FL [38]. Increased survival, due to the use of rituximab, has changed disease course to a more indolent one, affording time to define the effect of IFN-α treatment of aggressive lymphomas as part of an induction and maintenance strategy [39]. However, these studies are mostly single arm, due to the difficulty in obtaining a large sample size related to high mortality in these high-risk populations.

Table 2 Milestones of IFNs use for B cell malignancies

IFN-α-targeted immunocytokines in B cell lymphoma and myeloma

Although higher doses of IFN-α demonstrate greater anti-tumor activity, its significant systemic toxicities result in a very narrow therapeutic index (low maximum tolerated dose vs high optimal therapeutic dose). To address this limitation, several strategies have been explored to selectively deliver IFN-α to the tumor itself, including: (1) immunocytokines; (2) genetically modified DCs expressing IFN-α; (3) viral and other tumor-targeting vectors encoding IFN-α [40,41,42,43]; and (4) vectors encoding pattern recognition receptor agonists delivered directly into tumor microenvironment. One major strategy currently under pre-clinical development aims to target IFN-α to specific cell populations (such as malignant cells or specific types of leukocytes) by conjugating IFN-α to mAbs to generate antibody-based IFN-α fusion proteins, also called immunocytokines or immunoconjugates.

The potential benefit of an immunocytokine approach can be explained in part by mAb-induced target-specific cell death mediated via several indirect mechanisms: (1) immune effector cell-mediated antibody-dependent cellular cytotoxicity (ADCC); (2) complement-mediated cytotoxicity (CDC); (3) restoring immune effector cell function; and (4) direct mechanisms such as caspase-dependent apoptosis (Fig. 1). Indeed, the anti-CD38 mAbs inhibit immunosuppression exerted by regulatory T cells in MM [44,45,46] in addition to inducing myeloma cell death via lysosomal-associated and apoptotic pathways, which can be further enhanced by immunomodulatory drugs (IMiDs) [47]. Anti-CD38 mAbs may also inhibit MM-activated CD38+ pDC precursors [48] and/or restore DC maturation and presentation of tumor antigens, thereby further enhancing anti-tumor immunity. The addition of IFN-α was reported to augment ADCC by therapeutic mAbs both in vitro and in vivo [49, 50]. Specifically, mAb-mediated ADCC can be enhanced by IFN-α in the 3 ways: (1) enhancement of total target–mAb–effector binding by increasing tumor-associated antigen expression on tumor cells, as evidenced by in vitro studies showing that IFN-α induces CD20 upregulation on malignant B cells [51]; (2) activation of immune cells either directly, as IFN-α is a strong stimulus of NK cell activity, or indirectly through IFN-α-mediated upregulation of NKG2D ligands, which bind to co-stimulatory natural-killer group 2, member D (NKG2D) receptors expressed by NK cells, CD8 T cells, γδ T cells and macrophage [52]; and (3) blocking of effector cell ‘inhibitory’ signals, which remains largely unexplored. Additionally, type I IFN-containing immunocytokines can also target the tumor microenvironment by specifically binding to epidermal growth factor receptor on CTLs [53]. Taken together, cell-specific responses to tumor-targeting IFN-α-containing-mAbs relate to its sensitivity to IFN-α, the specific mAbs used, as well as the expression and density of the targeted tumor-associated antigens.

Fig. 1
figure 1

Enhancement of anti-tumor immunity by antibody-targeted IFN-α in B cell malignancies. Antibody-IFN-α fusion proteins are given by intravenous administration. The delivery of concentrated quantities of IFN-α to malignant sites is facilitated by tumor specific mAbs. Three potentially important mechanisms used by antibody-IFN-α fusion proteins to kill targeted tumor cells are: (1) IFN-αR mediated signals, i.e., IFN-α binds to membrane receptor IFN-αR expressed on tumor cells and activates downstream pathways to induce apoptosis; (2) IFN-α internalization, i.e., after mAb-IFN-α fusion proteins are internalized, IFN-α is released within cancer cells; (3) enhancing Fc receptor mediated ADCC, i.e., IFN-α augments ADCC exerted by mAbs through binding to the membrane receptor IFN-αR expressed on effector cells. E effector cells including NK cells, γδ T cells, macrophages and dendritic cells, B malignant B cells, IFN interferon, sIFN-αR soluble interferon alpha receptor, mAb monoclonal antibody, ADCC antibody-dependent cell-mediated cytotoxicity

Prior to the discovery of anti-CD20 mAbs, anti-tumor cytotoxic effect of mAbs was limited. In 1984, the idea of using mAbs to deliver IFNs into specific cellular compartments was first proposed in a human cancer model to exploit the anti-Epstein-Barr viral and anti-proliferative effects of IFNs [54]. In 1993, the anti-tumor activity of an immunoconjugate comprising natural IFN-α bound to a mAb specific for a human breast epithelial membrane mucin was studied in a xenograft tumor mouse model [20]. This study highlighted the potential feasibility of antibody-based IFN-α fusion proteins. Since then, the introduction of newer and highly potent mAbs (such as rituximab/anti-CD20, daratumumab/anti-CD38, elotuzumab/anti-SLAMF7) has renewed interest in the development of IFN-α-targeted immunocytokines. Pre-clinical studies now evaluating anti-CD20-IFN-α and anti-CD20-IFN-β against B cell lymphoma, as well as anti-CD138-IFN-α against myeloma [17, 55,56,57]. Genetically engineered anti-CD20-IFN-α fusion proteins exert direct cytotoxicity and overcome CD20 mAb resistance in mice bearing B-cell lymphoma xenografts [58]. In MM, anti-CD138-IFN-α fusion proteins in combination with bortezomib resulted in synergistic cytotoxicity in a MM mouse model [59]. These preclinical studies form the rationale for the subsequent clinical trials [60]. Phase I clinical evaluation of anti-CD20-IFN-α to treat B-cell lymphomas (ClinicalTrials.gov Identifier NCT02519270) has been initiated and is still ongoing (Table 3).

Table 3 Current trends in human monoclonal antibody fusion proteins targeting IFNs in B cell malignancies

In future, the ability to define patients who respond optimally to IFN-α-based immunotherapies is a central goal in cancer immunotherapy. Patients with B-cell lymphomas and MM are often immunocompromised, due to both the disease and its treatment. As IFN-α acts through activation of the immune system, a compromised immune system may limit IFN-α’s efficacy. In our opinions, IFN-α-based immunotherapies may benefit the following subpopulations of patients: (1) patients who have responded to intensive chemotherapy and stem cell transplantation, whose response may be deepened and prolonged by IFN-α-based immunotherapy; (2) patients with very early stage and/or indolent disease, limited tumor burden, and an intact immune response; (3) patients with robust anti-viral immunity, which can be reprogrammed to target cancer instead; and (4) patients with highly detectable proportions of circulating immune effector cells. The search continues for other potential biomarkers of response to IFN-α-based therapies, while genome-wide gene expression profiling (GEP) has in recent years emerged as a powerful tool. Taken rheumatoid arthritis for example, GEP revealed that pharmacodynamic differences in anti-CD20 mAb response very closely correlate to type-I IFN response gene activity [61]. Specifically, the increased expression of a set of 6 IFN response genes (RSAD2, IFI44, IFI44L, HERC5, LY6E and Mx1) was associated with a good response to mAb-based immunotherapy while higher baseline levels of type I IFNs may predict for lack of response to anti-CD20 mAbs.

Conclusions

The unique and multi-faceted anti-tumor mechanism of mAb-targeted IFN-α-based immunotherapy makes it a very promising agent for treatment of B cell malignancies. Moving forward, in vitro and in vivo preclinical studies are needed to further evaluate the therapeutic efficacy of mAb-targeted IFN-α-based immunotherapies both as monotherapy and in combination with other MM therapies including proteasome inhibitors, immunomodulatory drugs, and glucocorticoids. The following questions can be examined in preclinical models: (1) the relative anti-MM activity of intrinsic INF-α and anti-IFN-α mAb; (2) the reduction of tumor burden, including malignant stem cells, triggered by anti-IFN-α mAb compared to mAb alone; (3) impact of IFN-α on expression of tumor associated antigens (either on tumor cells or cells the within immune regulatory network) targeted by mAbs: by increasing the expression of CD20 on malignant B cells [51], anti-CD20-IFN-α has shown promising anti-tumor activity in patients who were unresponsive to anti-CD20 mAb-containing regimens; (4) tolerability of mAb-targeted IFN-α-based immunotherapies compared to systemic administration of IFN-α; and (5) whether mAb therapies modulate the IFN pathway. Additional studies are required to determine the optimal doses, schedules, and sequence of mAb-targeted IFN-α-based immunotherapies. Current research provides a strong rational for the early clinical evaluation of these agents. Ultimately, the clinical utility of these targeted IFN-based approaches will need to be validated in multicenter, randomized-controlled prospective studies.

Abbreviations

mAb:

monoclonal antibody

FDA:

Food and Drug Administration

MM:

multiple myeloma (MM)

SLAMF7:

SLAM Family Member

PFS:

progressive-free survival

CSCs:

cancer stem cells

MRD:

minimal residual disease

IFN:

interferons

IFN-α:

Interferon-alpha

CTCL:

cutaneous T cell lymphoma

DCs:

dendritic cells

IPCs:

IFN-producing cells

IFN-AR:

IFN-α receptor

STAT:

signal transducers and activators of transcription

CTL:

cytotoxic CD8+ T cell

Th1:

T helper-1

PD-1:

programmed cell death-1

NHL:

non-Hodgkin’s lymphoma

FL:

follicular lymphoma

ADCC:

antibody-dependent cellular cytotoxicity

CDC:

complement-mediated cytotoxicity

IMiDs:

immunomodulatory drugs

NKG2D:

natural-killer group 2, member D

GEP:

gene expression profiling

References

  1. Tai YT, Dillon M, Song W, Leiba M, Li XF, Burger P, Lee AI, Podar K, Hideshima T, Rice AG, van Abbema A, Jesaitis L, Caras I, Law D, Weller E, Xie W, et al. Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu. Blood. 2008;112:1329–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Richardson PG, Jagannath S, Moreau P, Jakubowiak AJ, Raab MS, Facon T, Vij R, White D, Reece DE, Benboubker L, Zonder J, Tsao LC, Anderson KC, Bleickardt E, Singhal AK, Lonial S, et al. Elotuzumab in combination with lenalidomide and dexamethasone in patients with relapsed multiple myeloma: final phase 2 results from the randomised, open-label, phase 1b-2 dose-escalation study. Lancet Haematol. 2015;2:e516–27.

    Article  PubMed  Google Scholar 

  3. Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, Hansson M, Minnema MC, Lassen U, Krejcik J, Palumbo A, van de Donk NW, Ahmadi T, Khan I, Uhlar CM, Wang J, Sasser AK, et al. Targeting CD38 with Daratumumab monotherapy in multiple myeloma. N Engl J Med. 2015;373:1207–19.

    Article  CAS  PubMed  Google Scholar 

  4. Palumbo A, Chanan-Khan A, Weisel K, Nooka AK, Masszi T, Beksac M, Spicka I, Hungria V, Munder M, Mateos MV, Mark TM, Qi M, Schecter J, Amin H, Qin X, Deraedt W, et al. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N Engl J Med. 2016;375:754–66.

    Article  CAS  PubMed  Google Scholar 

  5. Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci. 1957;147:258–67.

    Article  CAS  PubMed  Google Scholar 

  6. Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, Shah NK, Langer JA, Sheikh F, Dickensheets H, Donnelly RP. IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex. Nat Immunol. 2003;4:69–77.

    Article  CAS  PubMed  Google Scholar 

  7. Goeddel DV, Yelverton E, Ullrich A, Heyneker HL, Miozzari G, Holmes W, Seeburg PH, Dull T, May L, Stebbing N, Crea R, Maeda S, McCandliss R, Sloma A, Tabor JM, Gross M, et al. Human leukocyte interferon produced by E. coli is biologically active. Nature. 1980;287:411–6.

    Article  CAS  PubMed  Google Scholar 

  8. http://www.gene.com/media/press-releases/4174/1981-10-22/the-first-successful-expression-of-immun. Accessed 17 Jan 2017.

  9. Grever MR. How I treat hairy cell leukemia. Blood. 2010;115:21–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bunn PA Jr, Foon KA, Ihde DC, Longo DL, Eddy J, Winkler CF, Veach SR, Zeffren J, Sherwin S, Oldham R. Recombinant leukocyte A interferon: an active agent in advanced cutaneous T-cell lymphomas. Ann Intern Med. 1984;101:484–7.

    Article  PubMed  Google Scholar 

  11. Oken MM. New agents for the treatment of multiple myeloma and non-Hodgkin lymphoma. Cancer. 1992;70(4 Suppl):946–8.

    CAS  PubMed  Google Scholar 

  12. Adolf GR, Haas OA, Fischer P, Swetly P. Spontaneous production of alpha- and beta-interferon in human lymphoblastoid and lymphoma cell lines. Arch Virol. 1982;72:169–77.

    Article  CAS  PubMed  Google Scholar 

  13. Terawaki S, Chikuma S, Shibayama S, Hayashi T, Yoshida T, Okazaki T, Honjo T. IFN-alpha directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J Immunol. 2011;186:2772–9.

    Article  CAS  PubMed  Google Scholar 

  14. An G, Acharya C, Feng X, Wen K, Zhong M, Zhang L, Munshi NC, Qiu L, Tai YT, Anderson KC. Osteoclasts promote immune suppressive microenvironment in multiple myeloma: therapeutic implication. Blood. 2016;128:1590–603.

    Article  CAS  PubMed  Google Scholar 

  15. Gorgun G, Samur MK, Cowens KB, Paula S, Bianchi G, Anderson JE, White RE, Singh A, Ohguchi H, Suzuki R, Kikuchi S, Harada T, Hideshima T, Tai YT, Laubach JP, Raje N, et al. Lenalidomide enhances immune checkpoint blockade-induced immune response in multiple myeloma. Clin Cancer Res. 2015;21:4607–18.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Yang Y, Shaffer AL 3rd, Emre NC, Ceribelli M, Zhang M, Wright G, Xiao W, Powell J, Platig J, Kohlhammer H, Young RM, Zhao H, Yang Y, Xu W, Buggy JJ, Balasubramanian S, et al. Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma. Cancer Cell. 2012;21:723–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Huang TH, Chintalacharuvu KR, Morrison SL. Targeting IFN-alpha to B cell lymphoma by a tumor-specific antibody elicits potent antitumor activities. J Immunol. 2007;179:6881–8.

    Article  CAS  PubMed  Google Scholar 

  18. Maeda S, Wada H, Naito Y, Nagano H, Simmons S, Kagawa Y, Naito A, Kikuta J, Ishii T, Tomimaru Y, Hama N, Kawamoto K, Kobayashi S, Eguchi H, Umeshita K, Ishii H, et al. Interferon-alpha acts on the S/G2/M phases to induce apoptosis in the G1 phase of an IFNAR2-expressing hepatocellular carcinoma cell line. J Biol Chem. 2014;289:23786–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mullally A, Bruedigam C, Poveromo L, Heidel FH, Purdon A, Vu T, Austin R, Heckl D, Breyfogle LJ, Kuhn CP, Kalaitzidis D, Armstrong SA, Williams DA, Hill GR, Ebert BL, Lane SW. Depletion of Jak2V617F myeloproliferative neoplasm-propagating stem cells by interferon-alpha in a murine model of polycythemia vera. Blood. 2013;121:3692–702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ozzello L, De Rosa CM, Blank EW, Cantell K, Ceriani RL, Habif DV Sr. The use of natural interferon alpha conjugated to a monoclonal antibody anti mammary epithelial mucin (Mc5) for the treatment of human breast cancer xenografts. Breast Cancer Res Treat. 1993;25:265–76.

    Article  CAS  PubMed  Google Scholar 

  21. Grander D. How does interferon-alpha exert its antitumour activity in multiple myeloma? Acta Oncol. 2000;39:801–5.

    Article  CAS  PubMed  Google Scholar 

  22. Minami R, Muta K, Ilseung C, Abe Y, Nishimura J, Nawata H. Interleukin-6 sensitizes multiple myeloma cell lines for apoptosis induced by interferon-alpha. Exp Hematol. 2000;28:244–55.

    Article  CAS  PubMed  Google Scholar 

  23. Puthier D, Thabard W, Rapp M, Etrillard M, Harousseau J, Bataille R, Amiot M. Interferon alpha extends the survival of human myeloma cells through an upregulation of the Mcl-1 anti-apoptotic molecule. Br J Haematol. 2001;112:358–63.

    Article  CAS  PubMed  Google Scholar 

  24. Tai YT, Acharya C, An G, Moschetta M, Zhong MY, Feng X, Cea M, Cagnetta A, Wen K, van Eenennaam H, van Elsas A, Qiu L, Richardson P, Munshi N, Anderson KC. APRIL and BCMA promote human multiple myeloma growth and immunosuppression in the bone marrow microenvironment. Blood. 2016;127:3225–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mandelli F, Avvisati G, Amadori S, Boccadoro M, Gernone A, Lauta VM, Marmont F, Petrucci MT, Tribalto M, Vegna ML, et al. Maintenance treatment with recombinant interferon alfa-2b in patients with multiple myeloma responding to conventional induction chemotherapy. N Engl J Med. 1990;322:1430–4.

    Article  CAS  PubMed  Google Scholar 

  26. Durie BG, Russell DH, Salmon SE. Reappraisal of plateau phase in myeloma. Lancet. 1980;2:65–8.

    Article  CAS  PubMed  Google Scholar 

  27. Essers MA, Offner S, Blanco-Bose WE, Waibler Z, Kalinke U, Duchosal MA, Trumpp A. IFNalpha activates dormant haematopoietic stem cells in vivo. Nature. 2009;458:904–8.

    Article  CAS  PubMed  Google Scholar 

  28. Preudhomme C, Guilhot J, Nicolini FE, Guerci-Bresler A, Rigal-Huguet F, Maloisel F, Coiteux V, Gardembas M, Berthou C, Vekhoff A, Rea D, Jourdan E, Allard C, Delmer A, Rousselot P, Legros L, et al. Imatinib plus peginterferon alfa-2a in chronic myeloid leukemia. N Engl J Med. 2010;363:2511–21.

    Article  CAS  PubMed  Google Scholar 

  29. Gresser I, Coppey Y, Falcoff E, Fontaine D. Interferon and murine leukemia. I. Inhibitory effect of interferon on development of Friend leukemia in mice. Proc Soc Exp Biol Med. 1967; 124:84–91.

    Article  CAS  PubMed  Google Scholar 

  30. Strander H, Adamson U, Aparisi T, Broström LÅ, Cantell K, Einhorn S, Hall K, Ingimarsson S, Nilsonne U, Söderberg G. Adjuvant Interferon Treatment of Human Osteosarcoma. Springer, Berlin Heidelberg. 1979:34(6):877–80.

    Google Scholar 

  31. Merigan TC, Sikor K, Breeden JH, Levy R, Rosenberg SA. Preliminary Observations on the Effect of Human Leukocyte Interferon in Non-Hodgkin's Lymphoma. N Engl J Med. 1978;299:1449–53.

    Article  CAS  PubMed  Google Scholar 

  32. KA Foon, MS Roth, BP Jr. Interferon therapy of non-Hodgkin's lymphoma. Cancer. 1987;59(S3):601–4.

    Article  CAS  PubMed  Google Scholar 

  33. Hansen RM, Borden EC. Current status of interferons in the treatment of cancer. Oncology. 1992; 6(11):19–24.

    CAS  PubMed  Google Scholar 

  34. Foon KA. Biological response modifiers: the new immunotherapy. Cancer Res. 1989;49:1621–39.

    CAS  PubMed  Google Scholar 

  35. Ziegler-Heitbrock HW, Thiel E. Recombinant IFN-alpha in lymphomas. J Invest Dermatol. 1990;95(6 Suppl):213S–5S.

    Article  CAS  PubMed  Google Scholar 

  36. Mellstedt H, Aahre A, Bjorkholm M, Cantell K, Holm G, Johansson B, Strander H. Interferon therapy in myelomatosis. Lancet. 1979;2:697.

    CAS  PubMed  Google Scholar 

  37. Ludwig H, Fritz E. Interferon in multiple myeloma–summary of treatment results and clinical implications. Acta Oncologica. 2000;39(7):815–21.

    Article  CAS  PubMed  Google Scholar 

  38. Salles G, Mounier N, de Guibert S, Morschhauser F, Doyen C, Rossi JF, Haioun C, Brice P, Mahé B, Bouabdallah R. Rituximab combined with chemotherapy and interferon in follicular lymphoma patients: results of the GELA-GOELAMS FL2000 study. Blood. 2008;112(13):4824.

    Article  CAS  PubMed  Google Scholar 

  39. Geisler CH, Kolstad A, Laurell A, Andersen NS, Pedersen LB, Jerkeman M, Eriksson M, Nordström M, Kimby E, Boesen AM, Kuittinen O, Lauritzsen GF, Nilsson-Ehle H, Ralfkiær E, Akerman M, Ehinger M, Sundström C, Langholm R, Delabie J, Karjalainen-Lindsberg ML, Brown P, Elonen E. Long-term progression-free survival of mantle cell lymphoma after intensive front-line immunochemotherapy with in vivo–purged stem cell rescue: a nonrandomized phase 2 multicenter study by the Nordic Lymphoma Group. Blood. 2008;112:2687–93.

  40. Calabro ML, Gasperini P, Di Gangi IM, Indraccolo S, Barbierato M, Amadori A, Chieco-Bianchi L. Antineoplastic activity of lentiviral vectors expressing interferon-alpha in a preclinical model of primary effusion lymphoma. Blood. 2009;113:4525–33.

    Article  CAS  PubMed  Google Scholar 

  41. Dreno B, Urosevic-Maiwald M, Kim Y, Guitart J, Duvic M, Dereure O, Khammari A, Knol AC, Derbij A, Lusky M, Didillon I, Santoni AM, Acres B, Bataille V, Chenard MP, Bleuzen P, et al. TG1042 (Adenovirus-interferon-gamma) in primary cutaneous B-cell lymphomas: a phase II clinical trial. PLoS ONE. 2014;9:e83670.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Dummer R, Eichmuller S, Gellrich S, Assaf C, Dreno B, Schiller M, Dereure O, Baudard M, Bagot M, Khammari A, Bleuzen P, Bataille V, Derbij A, Wiedemann N, Waterboer T, Lusky M, et al. Phase II clinical trial of intratumoral application of TG1042 (adenovirus-interferon-gamma) in patients with advanced cutaneous T-cell lymphomas and multilesional cutaneous B-cell lymphomas. Mol Ther. 2010;18:1244–7.

    Article  CAS  PubMed Central  Google Scholar 

  43. Accart N, Urosevic-Maiwald M, Dummer R, Bataille V, Kehrer N, Niculescu C, Limacher JM, Chenard MP, Bonnefoy JY, Rooke R. Lymphocytic infiltration in the cutaneous lymphoma microenvironment after injection of TG1042. J Transl Med. 2013;11:226.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Feng X, Zhang L, Acharya C, An G, Wen K, Qiu L, Munshi NC, Tai YT, Anderson KC. Targeting CD38 suppresses induction and function of T regulatory cells to mitigate immunosuppression in multiple myeloma. Clin Cancer Res. 2017. doi:10.1158/1078-0432.CCR-16-3192.

  45. Tai YT, Anderson KC. A new era of immune therapy in multiple myeloma. Blood. 2016;128:318–9.

    Article  CAS  PubMed  Google Scholar 

  46. Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, Syed K, Liu K, van de Donk NW, Weiss BM, Ahmadi T, Lokhorst HM, Mutis T, Sasser AK. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016;128:384–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jiang H, Acharya C, An G, Zhong M, Feng X, Wang L, Dasilva N, Song Z, Yang G, Adrian F, Qiu L, Richardson P, Munshi NC, Tai YT, Anderson KC. SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia. 2016;30:399–408.

    Article  CAS  PubMed  Google Scholar 

  48. Bueno C, Montes R, Martin L, Prat I, Hernandez MC, Orfao A, Menendez P. NG2 antigen is expressed in CD34+ HPCs and plasmacytoid dendritic cell precursors: is NG2 expression in leukemia dependent on the target cell where leukemogenesis is triggered? Leukemia. 2008;22:1475–8.

    Article  CAS  PubMed  Google Scholar 

  49. Flieger D, Spengler U, Beier I, Kleinschmidt R, Hoff A, Varvenne M, Sauerbruch T, Schmidt-Wolf I. Enhancement of antibody dependent cellular cytotoxicity (ADCC) by combination of cytokines. Hybridoma. 1999;18:63–8.

    Article  CAS  PubMed  Google Scholar 

  50. Eisenthal A, Cameron RB, Rosenberg SA. Induction of antibody-dependent cellular cytotoxicity in vivo by IFN-alpha and its antitumor efficacy against established B16 melanoma liver metastases when combined with specific anti-B16 monoclonal antibody. J Immunol. 1990;144:4463–71.

    CAS  PubMed  Google Scholar 

  51. Sivaraman S, Venugopal P, Ranganathan R, Deshpande CG, Huang X, Jajeh A, Gregory SA, O’Brien T, Preisler HD. Effect of interferon-alpha on CD20 antigen expression of B-cell chronic lymphocytic leukemia. Cytokines Cell Mol Ther. 2000;6:81–7.

    Article  CAS  PubMed  Google Scholar 

  52. Zhang C, Niu J, Zhang J, Wang Y, Zhou Z, Zhang J, Tian Z. Opposing effects of interferon-alpha and interferon-gamma on the expression of major histocompatibility complex class I chain-related A in tumors. Cancer Sci. 2008;99:1279–86.

    Article  CAS  PubMed  Google Scholar 

  53. Yang X, Zhang X, Fu ML, Weichselbaum RR, Gajewski TF, Guo Y, Fu YX. Targeting the tumor microenvironment with interferon-beta bridges innate and adaptive immune responses. Cancer Cell. 2014;25:37–48.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Alkan SS, Miescher-Granger S, Braun DG, Hochkeppel HK. Antiviral and antiproliferative effects of interferons delivered via monoclonal antibodies. J Interferon Res. 1984;4:355–63.

    Article  CAS  PubMed  Google Scholar 

  55. Trinh KR, Vasuthasawat A, Steward KK, Yamada RE, Timmerman JM, Morrison SL. Anti-CD20-interferon-beta fusion protein therapy of murine B-cell lymphomas. J Immunother. 2013;36:305–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Yoo EM, Trinh KR, Tran D, Vasuthasawat A, Zhang J, Hoang B, Lichtenstein A, Morrison SL. Anti-CD138-targeted interferon is a potent therapeutic against multiple myeloma. J Interferon Cytokine Res. 2015;35:281–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Xuan C, Steward KK, Timmerman JM, Morrison SL. Targeted delivery of interferon-alpha via fusion to anti-CD20 results in potent antitumor activity against B-cell lymphoma. Blood. 2010;115:2864–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Vega GG, Franco-Cea LA, Huerta-Yepez S, Mayani H, Morrison SL, Bonavida B, Vega MI. Overcoming rituximab drug-resistance by the genetically engineered anti-CD20-hIFN-alpha fusion protein: direct cytotoxicity and synergy with chemotherapy. Int J Oncol. 2015;47:1735–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Vasuthasawat A, Yoo EM, Trinh KR, Lichtenstein A, Timmerman JM, Morrison SL. Targeted immunotherapy using anti-CD138-interferon alpha fusion proteins and bortezomib results in synergistic protection against multiple myeloma. MAbs. 2016;8:1386–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Pogue SL, Taura T, Bi M, Yun Y, Sho A, Mikesell G, Behrens C, Sokolovsky M, Hallak H, Rosenstock M, Sanchez E, Chen H, Berenson J, Doyle A, Nock S, Wilson DS. Targeting Attenuated Interferon-α to Myeloma Cells with a CD38 Antibody Induces Potent Tumor Regression with Reduced Off-Target Activity. PLoS One. 2016;11(9):e0162472.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Verweij CL, Vosslamber S. New insight in the mechanism of action of rituximab: the interferon signature towards personalized medicine. Discov Med. 2011;12:229–36.

    PubMed  Google Scholar 

Download references

Authors’ contributions

LZ review design; literature review; drafting the primary manuscript. Y-TT review design, critically edit the manuscript. MHZG table design; L-GQ literature review. KCA critically evaluated and edited the manuscript. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

K.C.A. serves on advisory boards to Onyx, Celgene, Gilead, and is a scientific founder of Acetylon, Oncopep, and C4 Therapeutics. The authors declare that they have no competing interests.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analyzed.

Funding

The Research Development Fund for Hematological Neoplasm from Chinese Anti-Cancer Association 312160342 and the National Natural Science Foundation of China for Young Scholars grants 81302148 (L.Z.). National Institutes of Health Grants RO1CA050947, RO1CA207237 and DF/HCC SPORE in Multiple Myeloma P50CA100707; K.C.A. is an American Cancer Society Clinical Research Professor.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Yu-Tzu Tai or Kenneth C. Anderson.

Additional file

40164_2017_81_MOESM1_ESM.xlsx

Additional file 1: Table S1. History of IFN-α treatment in human B cell malignancies. Abbreviations: IFN, interferon; ND, newly diagnosed; RR, relapsed and/or refractory; MCL, mantel cell lymphoma; SLL, small lymphocytic lymphoma; FL, follicular lymphoma; DLBCL, diffuse large B cell lymphoma; NHL, non-Hodgkin lymphoma; MALT, mucosa-associated lymphoid tissue; HL, Hodgkin lymphoma; pcMZL, primary cutaneous marginal zone lymphoma; CLL, chronic lymphocytic lymphoma; AITL, angioimmunoblastic T-cell lymphoma; PTCL, peripheral T cell lymphoma; DLCL, diffuse large cell lymphoma; MM, multiple myeloma; WM, Waldenstrom’s macroglobulinemia; NK, natural killer; LG, large granular; IL, interleukin; HDT, high dose treatment; ASCT, autologous stem cell transplant; ABSCT, autologous bone marrow or blood stem cell transplantation; PBSCT, peripheral blood stem cell transplantation; CHOP, cyclophosphamide, vincristine, doxorubicin, and prednisone; UA, unavailable; *IFN Dose: high-dose IFN (5 MIU/m2 daily), low-dose (3 MIU/m2/day, 3 times a week), intermediate-dose, between high and low doses.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, L., Tai, YT., Ho, M.Z.G. et al. Interferon-alpha-based immunotherapies in the treatment of B cell-derived hematologic neoplasms in today’s treat-to-target era. Exp Hematol Oncol 6, 20 (2017). https://doi.org/10.1186/s40164-017-0081-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-017-0081-6

Keywords