Chemotherapy: Open Access

Chemotherapy: Open Access
Open Access

ISSN: 2167-7700

Commentary - (2016) Volume 5, Issue 2

Targeting Human β-Microglobulin with Monoclonal Antibodies in Multiple Myeloma - A Potential in Treatment

Mingjun Zhang, Jin He and Jing Yang*
1Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
2Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, USA
3Cancer Research Institution, Guangzhou Medical University, Guangzhou, China
*Corresponding Author: Jing Yang, Division of Cancer Medicine, Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Texas, USA, Tel: 713-563-0357, Fax: 713-745-1179 Email:

Abstract

Multiple myeloma (MM) is a clonal plasma cell neoplasm that utilizes bone marrow microenvironment for survival and proliferation [1-3]. However, current therapies could rarely cure MM. The relapse or refractory aspect of the disease is commonly seen in MM patients, especially among patients with high-risk MM. In past decades, targeted immunotherapy with monoclonal antibodies (mAbs) emerged as a major new treatment modality that offered great benefits for MM patients [4]. Different approaches, aimed at finding potential mAbbased therapeutics for this disease including identification of alternative, or novel, target antigens [5], conjugation of mAbs with classic or novel drugs [6], and generation of chimeric antigen receptor T cells with specific mAbs [7], have been developed by scientists. Recently, our group has generated the mAbs that work directly against human β2-microglobulin (β2M) both in vitro and in the mouse experiments, and has demonstrated that β2M is a potential target for MM treatment [8].

Keywords: Monoclonal, Antibodies, Neoplasm, Immunotherapy, Multiple myeloma

Introduction

Multiple myeloma (MM) is a clonal plasma cell neoplasm that utilizes bone marrow microenvironment for survival and proliferation [1-3]. However, current therapies could rarely cure MM. The relapse or refractory aspect of the disease is commonly seen in MM patients, especially among patients with high-risk MM. In past decades, targeted immunotherapy with monoclonal antibodies (mAbs) emerged as a major new treatment modality that offered great benefits for MM patients [4]. Different approaches, aimed at finding potential mAbbased therapeutics for this disease including identification of alternative, or novel, target antigens [5], conjugation of mAbs with classic or novel drugs [6], and generation of chimeric antigen receptor T cells with specific mAbs [7], have been developed by scientists. Recently, our group has generated the mAbs that work directly against human β2-microglobulin (β2M) both in vitro and in the mouse experiments, and has demonstrated that β2M is a potential target for MM treatment [8].

Human β2M is part of major histocompatibility complex (MHC) class I molecules [9], that is involved in the presentation of peptide antigens to immune cells. Elevated β2M levels can be observed in patients with MM or other hematological malignancies, and this molecule has served as one of the key prognosis indicators in MM [10,11]. Using human-like mouse models, our research has demonstrated that anti-β2M mAbs have strong and direct apoptotic effects on MM (Figure 1A) and other hematological malignancies, with little toxicity towards normal tissues and cells [12]. The anti-β2M mAbs activate the c-Jun N-terminal kinases and inhibit extracellularsignal- regulated kinases and phosphatidylinositide 3-kinases/Akt (also known as protein kinase B). The mediated signaling pathways, and the mAbs, can recruit MHC class I molecules into and exclude receptors for growth factors, such as IL-6 and IGF-1, from lipid rafts [12,13]. Our results suggest that anti-β2M mAbs could be a novel therapeutic agent specifically targeting MM in a clinical setting.

chemotherapy-Schematic-representation

Figure 1: Schematic representation of the mechanistic actions of anti-β2M mAbs against MM cells. Anti-β2M mAbs induce MM cell death via (A) induction of MM cell apoptosis, and activation of (B) CDC and (C) ADCC. Lenalidomide could enhance anti-β2M mAbinduced ADCC activity by increasing the activity of NK cells. (D) Combination treatment of BTZ and anti-β2M mAbs overcomes drug resistance of BTZ by inhibiting BTZ-induced autophagy and increasing MM cell apoptosis.

In addition, enhancing antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activities is one of the most promising ways to improve the clinical efficacy of already-approved antibodies. This concept is now actively being examined in the clinic, especially in the field of hematological malignancy treatment [14]. Our recent studies show that anti-β2M mAbs effectively lysed MM cells via ADCC and CDC (Figure 1B and 1C). We examined the anti-MM activity of anti-β2M mAbs combined with lenalidomide, an immunomodulatory drug that has been widely used in the treatment of MM [15], and we found that lenalidomide potentiated the mAb-induced ADCC activity both in vitro and in vivo against MM cells by enhancing the killing activity of natural killer cells (Figure 1C) [16]. These findings provide a rationale for combining anti-β2M mAbs with lenalidomide to improve patient outcomes in MM.

Another standard regimen to treat MM patients is proteasome inhibitor-based chemotherapy. As an example, bortezomib (BTZ) is currently being used worldwide to treat MM and mantle cell lymphoma [17]. However, adverse effects and drug resistance are emerging as great challenges for its extended application [18]. We speculated about whether the addition of anti-β2M mAb treatment would indeed improve the efficacy of BTZ alone. Our investigations showed that the combination treatment offered a much higher anti- MM effects than either agent alone, and anti-β2M mAbs enhanced BTZ-induced apoptosis in MM cells and in mouse models. Mechanistic studies showed that anti-β2M mAbs could overcome BTZ resistance by inhibiting BTZ-induced nuclear factor kappa-light-chainenhancer of activated B cells (NF-κB) signaling and autophagy activation (Figure 1D) [19]. Thus, our studies provide a new insight in the development of anti-β2M mAbs and BTZ combination to overcome chemotherapy resistance in MM patients.

In summary, our results suggest that anti-β2M mAbs may be a more promising next-generation antibody-based immunotherapeutic agent for the treatment of MM. The clinical development of anti-β2M mAbs, both as a monotherapy or in combination with existing MM drugs, such as lenalidomide or BTZ, offers MM patients increased treatment options and improves overall patient outcome.

Acknowledgments

We thank Ms. Victoria M. Leyton at the University of Texas, MD Anderson Cancer Center (MDACC) for providing editorial assistance. This work was supported by the National Cancer Institute R01s (1R01CA190863 and 1R01CA193362; J. Yang), the American Cancer Society Research Scholar Grant (127337-RSG-15-069-01-TBG; J. Yang), the MDACC IRG-Basic Research (J. Yang), the Leukemia Research Foundation (J. Yang), the American Society of Hematology (J. Yang), and the National Natural Science Foundation of China (Grant No. 81470356; J. Yang). The authors have no competing financial interests.

References

  1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, et al. (2015) Global cancer statistics, 2012. CA Cancer J Clin 65: 87-108.
  2. Bluthgen MV, Besse B (2015) Second-line combination therapies in non-small cell lung cancer without known driver mutations. Eur Respir Rev 24: 582-593.
  3. Okamoto I, Takahashi T, Okamoto H, Nakagawa K, Watanabe K, et al. (2011) Single-agent gefitinib with concurrent radiotherapy for locally advanced non-small cell lung cancer harboring mutations of the epidermal growth factor receptor. Lung Cancer 72: 199-204.
  4. Russo A, Franchina T, Ricciardi GR, Picone A, Ferraro G, et al. (2015) A decade of EGFR inhibition in EGFR-mutated non-small cell lung cancer (NSCLC): Old successes and future perspectives. Oncotarget 6: 26814-26825.
  5. Chen YM (2013) Update of epidermal growth factor receptor-tyrosine kinase inhibitors in non-small-cell lung cancer. J Chin Med Assoc 76: 249-257.
  6. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, et al. (2009) Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361: 947-957.
  7. Han JY, Park K, Kim SW, Lee DH, Kim HY, et al. (2012) First-SIGNAL: first-line single-agent iressa versus gemcitabine and cisplatin trial in never-smokers with adenocarcinoma of the lung. J Clin Oncol 30: 1122-1128.
  8. Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, et al. (2010) Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 11: 121-128.
  9. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, et al. (2010) Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 362: 2380-2388.
  10. Zhou C, Wu YL, Chen G, Feng J, Liu XQ, et al. (2011) Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 12: 735-742.
  11. Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, et al. (2012) Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 13: 239-246.
  12. Sequist LV, Yang JC, Yamamoto N, O'Byrne K, Hirsh V, et al. (2013) Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 31: 3327-3334.
  13. Wu YL, Zhou C, Hu CP, Feng J, Lu S, et al. (2014) Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol 15: 213-222.
  14. Castellanos EH, Horn L (2015) Generations of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors: Perils and Progress. Curr Treat Options Oncol 16: 51.
  15. Chen H, Yao W, Chu Q, Han R, Wang Y, et al. (2015) Synergistic effects of metformin in combination with EGFR-TKI in the treatment of patients with advanced non-small cell lung cancer and type 2 diabetes. Cancer Lett 369: 97-102.
  16. Forde PM, Ettinger DS (2015) Managing acquired resistance in EGFR-mutated non-small cell lung cancer. Clin Adv Hematol Oncol 13: 528-532.
  17. Yu HA, Sima CS, Huang J, Solomon SB, Rimner A, et al. (2013) Local therapy with continued EGFR tyrosine kinase inhibitor therapy as a treatment strategy in EGFR-mutant advanced lung cancers that have developed acquired resistance to EGFR tyrosine kinase inhibitors. J Thorac Oncol 8: 346-351.
  18. McCloskey P, Balduyck B, Van Schil PE, Faivre-Finn C, O'Brien M (2013) Radical treatment of non-small cell lung cancer during the last 5 years. Eur J Cancer 49: 1555-1564.
  19. Ouyang WW, Su SF, Hu YX, Lu B, Ma Z, et al. (2014) Radiation dose and survival of patients with stage IV non-small cell lung cancer undergoing concurrent chemotherapy and thoracic three-dimensional radiotherapy: reanalysis of the findings of a single-center prospective study. BMC Cancer 14: 491.
  20. Slotman BJ, van Tinteren H, Praag JO, Knegjens JL, Sharouni SY, et al. (2015) Use of thoracic radiotherapy for extensive stage small-cell lung cancer: a phase 3 randomised controlled trial. Lancet 385: 36-42.
  21. Moschini I, Dell'Anna C, Losardo PL, Bordi P, D'Abbiero N, et al. (2015) Radiotherapy of non-small-cell lung cancer in the era of EGFR gene mutations and EGF receptor tyrosine kinase inhibitors. Future Oncol 11: 2329-2342.
  22. Tanaka T, Munshi A, Brooks C, Liu J, Hobbs ML, et al. (2008) Gefitinib radiosensitizes non-small cell lung cancer cells by suppressing cellular DNA repair capacity. Clin Cancer Res 14: 1266-1273.
  23. Ntaskagiannis D, Gogou P, Murray S, Sainis I, Briasoulis E, et al. (2015) The effect of EGFR mutation status in the outcome of radiotherapy in patients with locally advanced non-small cell lung cancer (NSCLC). Annals of Cancer Research 2: 4.
  24. Chang CC, Chi KH, Kao SJ, Hsu PS, Tsang YW, et al. (2011) Upfront gefitinib/erlotinib treatment followed by concomitant radiotherapy for advanced lung cancer: a mono-institutional experience. Lung Cancer 73: 189-194.
  25. Iyengar P, Kavanagh BD, Wardak Z, Smith I, Ahn C, et al. (2014) Phase II trial of stereotactic body radiation therapy combined with erlotinib for patients with limited but progressive metastatic non-small-cell lung cancer. J Clin Oncol 32: 3824-3830.
  26. Wu YL, Zhou C, Liam CK, Wu G, Liu X, et al. (2015) First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study. Ann Oncol 26: 1883-1889.
  27. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, et al. (2009) New response evaluation criteria in solid tumours: revised RECIST guideline Eur J Cancer 45: 228-247.
  28. Thongprasert S, Duffield E, Saijo N, Wu YL, Yang JC, et al. (2011) Health-related quality-of-life in a randomized phase III first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients from Asia with advanced NSCLC (IPASS). J Thorac Oncol 6: 1872-1880.
  29. Hay JL, Atkinson TM, Reeve BB, Mitchell SA, Mendoza TR, et al. (2014) Cognitive interviewing of the US National Cancer Institute's Patient-Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE). Qual Life Res 23: 257-269.
Citation: Zhang M, He J, Yang J (2016) Targeting Human β-Microglobulin with Monoclonal Antibodies in Multiple Myeloma - A Potential in Treatment. Chemo Open Access 5:190.

Copyright: © 2016 Zhang M, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top