International Journal of Pharmaceutical and Phytopharmacological Research
ISSN (Print): 2250-1029
ISSN (Online): 2249-6084
Publish with eIJPPR Submission
2023   Volume 13   Issue 3

A Comprehensive Review of Anti-CD20 Monoclonal Antibodies in Multiple Sclerosis

Ramdas Bhat1*, Preeti Shanbhag1, Ramakrishna Shabaraya2

 

1Department of Pharmacology, Srinivas College of Pharmacy, Mangalore, Karnataka, India.

2Srinivas College of Pharmacy, Mangalore, Karnataka, India.


ABSTRACT

Multiple sclerosis (MS) poses a complex challenge, impacting countless individuals worldwide. The disease's multifaceted pathogenesis has stymied researchers and clinicians. Yet, headway has been made in understanding its immunological roots, leading to novel therapies like Anti-CD20 Monoclonal Antibodies (mAbs) that target B cells. These mAbs offer a precise approach to immune modulation, holding the potential to arrest disease progression and reduce relapses. This review delves into the efficacy, mechanisms, and safety of Anti-CD20 mAbs in MS treatment. Clinical trials consistently demonstrate their effectiveness in lowering relapse rates, slowing disability progression, and enhancing patients' quality of life. While concerns about infections exist, safety data support their viability. Comparative analysis of mAbs like rituximab, ocrelizumab, ofatumumab, and ublituximab unveils their unique mechanisms and dosing regimens. For long-term success, treatment optimization is paramount, balancing sustained disease control with safety monitoring, personalization, and quality of life considerations. Combining mAbs with existing or emerging therapies shows promise. Looking ahead, precision medicine, cellular therapies, and digital health technologies promise to revolutionize MS care. This review equips clinicians, researchers, and stakeholders with a comprehensive understanding of Anti-CD20 mAbs potential in MS management and envisions an era of tailored therapies and innovation in MS treatment.

Key Words: Multiple sclerosis (MS), Anti-CD20 monoclonal antibodies (mAbs), Neuroinflammatory disease, FasL (Fas ligand), Rituximab


INTRODUCTION

Multiple sclerosis (MS) stands as one of the most enigmatic and challenging neuroinflammatory diseases, affecting millions of individuals worldwide [1]. With its intricate and multifaceted pathogenesis, MS has long presented a formidable clinical conundrum for researchers and clinicians alike [2, 3]. Over the years, considerable progress has been made in understanding the underlying immunological mechanisms driving the disease, leading to the development of novel therapeutic strategies aimed at taming the relentless immune assault on the central nervous system [4]. Among these groundbreaking approaches, the utilization of Anti-CD20 Monoclonal Antibodies (mAbs) has emerged as a transformative frontier in MS treatment [5, 6]. These agents, designed to selectively target CD20-expressing B cells, have revolutionized the therapeutic landscape by offering a more precise and personalized approach to immune modulation. By strategically depleting the pathogenic B cell population, Anti-CD20 mAbs have shown unprecedented potential in halting disease progression, reducing relapse rates, and ameliorating neurological disability [7].

This review article endeavors to provide a comprehensive and in-depth analysis of the efficacy of Anti-CD20 Monoclonal Antibodies in the treatment of multiple sclerosis. Through a meticulous examination of preclinical studies, pivotal clinical trials, and real-world evidence, we aim to unravel the mechanistic insights underpinning their therapeutic impact. By scrutinizing the accumulating body of evidence, we seek to elucidate the comparative effectiveness of Anti-CD20 mAbs against conventional MS therapies, providing a critical assessment of their position in the therapeutic armamentarium. Throughout this review, we will explore the multifaceted mechanisms of action of Anti-CD20 mAbs, including their profound influence on B cell depletion, subsequent modulation of other immune cell populations, and the intricate interplay with the neuroinflammatory milieu. Additionally, we will analyze the long-term outcomes and safety profiles of these agents, offering valuable insights into their durability and tolerability in clinical practice. As we embark on this journey through the realms of cutting-edge MS therapy, it is essential to highlight the evolving landscape of treatment paradigms. We will discuss the potential for combining Anti-CD20 mAbs with other disease-modifying therapies and the burgeoning concept of personalized medicine to optimize treatment outcomes for individual patients. By synthesizing the latest advancements and clinical experiences, this review aspires to equip clinicians, researchers, and healthcare stakeholders with a comprehensive understanding of the promises and challenges associated with Anti-CD20 Monoclonal Antibodies in multiple sclerosis management. As we harness the power of these revolutionary agents, we envision a future where individuals with MS can attain an improved quality of life and renewed hope for effective disease control.

Pathogenesis of multiple sclerosis

Multiple sclerosis (MS) is a multifaceted autoimmune disease characterized by a complex pathogenesis involving various immunological processes [8]. The disease's onset is triggered by a disruption in immune tolerance, leading to the activation of autoreactive T cells and B cells. In this context, the Fas-FasL signaling pathway plays a critical role. Activated autoreactive T cells infiltrate the central nervous system (CNS) through the blood-brain barrier (BBB) and initiate an inflammatory response against myelin, the protective sheath surrounding nerve fibers [9]. Subsequently, CD4+ T helper cells produce pro-inflammatory cytokines, stimulating microglia and macrophages to further contribute to neuroinflammation and demyelination. In parallel, CD8+ T cells target and destroy myelin-producing oligodendrocytes directly. The upregulation of Fas receptor expression on oligodendrocytes renders them susceptible to Fas ligand (FasL)-mediated apoptosis induced by infiltrating immune cells. This cascade of events leads to demyelination, neurodegeneration, and the formation of inflammatory lesions within the CNS, resulting in the hallmark clinical symptoms of MS. The heterogeneity of MS subtypes, such as relapsing-remitting MS (RRMS) and progressive MS (including primary progressive MS and secondary progressive MS), reflects the complexity and diversity of the underlying immunological dysregulation in the pathogenesis of this debilitating disease [10-12].

Mechanism of action of anti-CD20 monoclonal antibodies over B cell

Anti-CD20 Monoclonal Antibodies (mAbs) have a profound impact on B cells and their interaction with the Fas-FasL pathway [13]. CD20 is a surface protein predominantly expressed in B cells, and these mAbs are specifically designed to target and bind to CD20, leading to B cell depletion [14]. Upon administration of Anti-CD20 mAbs, they recognize and attach to CD20 molecules on the surface of B cells. This binding trigger various mechanisms that result in the reduction of B cell numbers [15, 16]. One of the main mechanisms is antibody-dependent cell-mediated cytotoxicity (ADCC). Natural killer (NK) cells, equipped with Fc receptors, recognize the bound Anti-CD20 mAbs and subsequently bind to the B cells. This interaction activates the NK cells, leading to the release of cytotoxic substances that induce apoptosis, causing targeted B cell death. Fas-FasL pathway, B cells express Fas (CD95), a receptor protein, on their surface. FasL (Fas ligand) is expressed on various immune cells, including activated T cells. The binding of FasL to Fas triggers a signaling cascade that induces apoptosis in the Fas-expressing cell. In some instances, Anti-CD20 mAbs can synergize with the Fas-FasL pathway to promote B cell apoptosis. This occurs when FasL-expressing immune cells, like activated T cells, recognize Anti-CD20 mAb-bound B cells. The FasL-Fas interaction amplifies the apoptotic signal in B cells, leading to further B cell depletion. The combination of Anti-CD20 mAbs' direct effects on B cells, such as ADCC, along with their potential to engage the Fas-FasL pathway, results in a potent and precise reduction of B cells in the context of autoimmune diseases and conditions where B cell dysregulation plays a significant role [17, 18]. By targeting CD20 and utilizing the Fas-FasL pathway, Anti-CD20 mAbs offer a promising therapeutic approach in various disorders, including multiple sclerosis, rheumatoid arthritis, and certain types of B cell lymphomas [18].

Clinical efficacy

Anti-CD20 monoclonal antibodies (mAbs) have been extensively evaluated in clinical trials to assess their clinical efficacy in treating both relapsing-remitting multiple sclerosis (RRMS) and primary progressive multiple sclerosis (PPMS) patients. The findings from these trials consistently demonstrate the remarkable effectiveness of anti-CD20 mAbs across key outcome measures [19, 20]. Clinical trials investigating anti-CD20 mAbs have shown a significant reduction in relapse rates when compared to placebo or other disease-modifying therapies. The ability of these mAbs to suppress B cells and modulate the immune response plays a pivotal role in mitigating the frequency and severity of disease exacerbations. Furthermore, anti-CD20 mAbs have a notable impact on disability progression, as evidenced by studies utilizing standardized disability scales like the Expanded Disability Status Scale (EDSS). Treatment with anti-CD20 mAbs slows down the progression of disability compared to placebo or comparator therapies, likely through their ability to reduce inflammatory activity and neurodegeneration. Magnetic resonance imaging (MRI) assessments reveal a consistent reduction in MRI lesion burden with the use of anti-CD20 mAbs compared to controls. This reduction provides evidence of the mAbs' capacity to suppress inflammatory processes and halt disease progression. Importantly, MS patients report improvements in their quality of life following treatment with anti-CD20 mAbs. Patient-reported outcome measures consistently indicate enhancements in everyday functioning, social activities, and emotional well-being, emphasizing the positive impact of anti-CD20 mAbs on patients’ lives [21-24].

Safety profile

The safety profile of anti-CD20 monoclonal antibodies (mAbs) in multiple sclerosis (MS) treatment is a critical consideration. While the risk of infections is a concern, the available evidence suggests that the overall incidence of serious infections is comparable to or only slightly higher than other disease-modifying therapies. Infusion reactions are relatively common, but most are mild to moderate in severity and can be managed with premedication and close monitoring [25-27]. Additionally, long-term follow-up studies demonstrate that immune system parameters tend to return to baseline levels over time after treatment discontinuation. Moreover, comparative safety data reveal subtle differences in the occurrence of specific adverse events among various anti-CD20 mAbs, providing valuable insights for tailoring treatment choices to individual patient needs. These findings collectively support the continued evaluation and utilization of anti-CD20 mAbs as a viable and safe therapeutic option for MS management [28-30].

Comparative analysis of anti-CD20 monoclonal antibodies [31-34]

Several anti-CD20 monoclonal antibodies are utilized in the treatment of multiple sclerosis, each distinguished by its unique mechanism of action, dose regimen, and frequency of administration as shown in Table 1.

 

 

Table 1. Various anti-CD20 monoclonal antibodies based on their mechanisms of action, dose regimens, and frequency of administration for the treatment of multiple sclerosis.

Anti-CD20 monoclonal antibodies

Mechanism

Dose regimen

Frequency of administration

Rituximab

B cell depletion by antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and induction of apoptosis.

Two 1000 mg IV infusions separated by two weeks.

Every 6 to 12 months

Ocrelizumab

 

B cell depletion through similar mechanisms as rituximab.

Initial: Two 300 mg IV infusions separated by two weeks, Subsequent: Single 600 mg IV infusions

Every six months after initial infusions

Ofatumumab

 

B cell depletion targeting distinct CD20 epitope compared to rituximab and ocrelizumab

Subcutaneous: Two 20mg injections two weeks apart for the first dose, followed by a single 20 mg injection after one week

Subsequent doses are given monthly after initial injections

Ublituximab

B cell depletion is designed to induce enhanced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity

Intravenous administration with varying intervals in the dosing regimen

Administered in cycles with varying intervals between cycles

 

Long-term outcomes and treatment optımızatıon

When dealing with the long-term outcomes and optimization of treatment involving anti-CD20 monoclonal antibodies (Table 1), several essential aspects come into play. It's crucial to assess how effectively these antibodies sustain disease control over extended periods, while also closely monitoring and managing potential safety concerns and risks that may arise in the long run. Determining the optimal treatment duration and frequency is key to striking the right balance between therapeutic benefits and potential side effects [35-37]. Tailoring treatment plans to suit individual patient needs, exploring combination therapies to enhance effectiveness, and assessing the impact on a patient's overall quality of life are vital considerations [38]. Additionally, evaluating the cost-effectiveness of long-term antibody treatment, establishing a systematic monitoring schedule, and actively involving patients in decision-making processes all contribute to achieving favorable and sustainable long-term outcomes [39].

Combınatıon therapy

The investigation of combining anti-CD20 monoclonal antibodies (mAbs) like ocrelizumab or rituximab with proven disease-modifying medicines or cutting-edge therapeutic methods is highlighted in the changing world of multiple sclerosis (MS) care. This section deftly negotiates the terrain of combining these powerful mAbs with already available drugs like interferons and glatiramer acetate or cutting-edge therapies like sphingosine-1-phosphate receptor modulators [40, 41]. The rationale, safety considerations, and potential synergistic benefits of these combination endeavors are meticulously scrutinized, encompassing considerations of immune modulation, targeted pathways, and cumulative therapeutic effects [42]. By unraveling mechanisms, interactions, and real-world data, this analysis unveils the harmonious interplay between diverse therapeutic elements. Additionally, exploring potential combinations with emerging immunomodulatory agents or novel approaches like cellular therapies adds another layer of complexity to the landscape. This comprehensive review contributes to a refined understanding of the intricate dynamics, equipping clinicians to optimize multiple sclerosis management by harnessing the potential of anti-CD20 mAbs in tandem with other drugs, all while anticipating the broader horizons of therapeutic combinations in the evolving MS treatment paradigm. This progressive avenue of combining therapies stands as a testament to the evolving nature of MS treatment strategies, where each synergy represents a step towards a harmonized and personalized approach to addressing the complexities of the disease [43-46].

Future dırectıons

Gazing ahead within the landscape of multiple sclerosis (MS) treatments, this section embraces the unfolding vistas of prospects that hold promise for refining therapeutic paradigms. From innovative approaches in drug delivery and precision medicine to the advent of advanced technologies like gene editing and personalized immunotherapies, the horizon is illuminated with potential breakthroughs [47].

Precision medicine is poised to ascend as a cornerstone of MS management, where individualized treatment plans are tailored based on genetic, biomarker, and clinical profiles [48]. The integration of genomic data, coupled with sophisticated bioinformatics, empowers clinicians to predict treatment responses and optimize interventions for enhanced efficacy. Emerging therapeutic avenues, such as cell-based therapies and gene editing, stand at the threshold of transformational change. The dawn of engineered immune cells and the manipulation of disease-associated genes offer the potential to recalibrate the immune system’s response, heralding a new era of targeted interventions with the potential to halt disease progression [49].

In parallel, digital health technologies cast a transformative spell, fostering real-time monitoring, patient engagement, and telemedicine, thereby amplifying disease management beyond traditional boundaries. Wearables, mobile apps, and data analytics converge to empower patients and clinicians alike, enabling timely adjustments to treatment regimens. The future of MS management transcends the confines of traditional therapeutics, embracing interdisciplinary collaboration, advanced technologies, and patient-centered care [50].

CONCLUSION

This comprehensive review highlights the transformational potential of anti-CD20 monoclonal antibodies (mAbs) in the field of multiple sclerosis (MS) care. These drugs exhibit outstanding efficacy in delaying disease progression, lowering relapse rates, and improving patient well-being because they target B cells through complex pathways. As we look to the future, digital health technology, cell-based medicines, and precision medicine show promising potential. This review not only gives doctors and researchers insights into clinical nuances and safety profiles, but it also predicts a time in the future when individualized approaches and innovation will transform the MS management landscape, ushering in a new era of scientific advancement and compassionate care.

Acknowledgments: I would like to thank Staffs and Management of Srinivas College of Pharmacy for their Support.

Conflict of interest: None

Financial support: None

Ethics statement: None

References

[1]    Reich DS, Lucchinetti CF, Calabresi PA. Multiple Sclerosis. Longo DL, editor. N Engl J Med. 2018;378(2):169-80.

[2]    Sadeghi M, Rahimi M, Poornoroz N, Jahromi FF. Patient satisfaction with hospital services after the implementation of the health system. Arch Pharm Pract. 2021;12(1):31-6.

[3]    Lampasona M, Pantaleo L. The Role of Pharmacies in Immunization Programs and Health Promotion. Arch Pharm Pract. 2022;13(2):62-5.

[4]    Dighriri IM, Aldalbahi AA, Albeladi F, Tahiri AA, Kinani EM, Almohsen RA, et al. An Overview of the History, Pathophysiology, and Pharmacological Interventions of Multiple Sclerosis. Cureus. 2023;15(1):e33242.

[5]    Arikawe O, Morrissey H, Ball P. Community pharmacy brief screening intervention to improve health outcomes for patients diagnosed with chronic diseases. J Adv Pharm Educ Res. 2022;12(3):1-8.

[6]    Ammoo AM, Al-Tamimi DJJ, Al-Mahroos MIA, Al-Tamimi MJJ, Ibraheem JJ. Pharmacokinetics of Fluconazole tablets administered to healthy subjects. J Adv Pharm Educ Res. 2021;11(2):92-9.

[7]    Nguyen AL, Gresle M, Marshall T, Butzkueven H, Field J. Monoclonal antibodies in the treatment of multiple sclerosis: emergence of B-cell-targeted therapies. Br J Pharmacol. 2017;174(13):1895-907.

[8]    Lemus HN, Warrington AE, Rodriguez M. Multiple Sclerosis. Neurol Clin. 2018;36(1):1-11.

[9]    Goverman J. Autoimmune T cell responses in the central nervous system. Nat Rev Immunol. 2009;9(6):393-407.

[10] Sato F, Omura S, Jaffe SL, Tsunoda I. Role of CD4+ T Cells in the Pathophysiology of Multiple Sclerosis. Mult Scler. 2016:41-69.

[11] Kaya T, Mattugini N, Liu L, Ji H, Cantuti-Castelvetri L, Wu J, et al. CD8+ T cells induce interferon-responsive oligodendrocytes and microglia in white matter aging. Nat Neurosci. 2022;25(11):1446-57.

[12] Höftberger R, Lassmann H. Inflammatory demyelinating diseases of the central nervous system. Neuropathology. 2018;263-83.

[13] Klineova S, Lublin FD. Clinical Course of Multiple Sclerosis. Cold Spring Harb Perspect Med. 2018;8(9):a028928.

[14] Volpe E, Sambucci M, Battistini L, Borsellino G. Fas–Fas Ligand: Checkpoint of T Cell Functions in Multiple Sclerosis. Front Immunol. 2016;7:382.

[15] Boross P, Leusen JH. Mechanisms of action of CD20 antibodies. Am J Cancer Res. 2012;2(6):676-90.

[16] Pavlasova G, Mraz M. The regulation and function of CD20: an “enigma” of B-cell biology and targeted therapy. Haematologica. 2020;105(6):1494-506.

[17] Casan JML, Wong J, Northcott MJ, Opat S. Anti-CD20 monoclonal antibodies: reviewing a revolution. Hum Vaccin Immunother. 2018;14(12):2820-41.

[18] Lo Nigro C, Macagno M, Sangiolo D, Bertolaccini L, Aglietta M, Merlano MC. NK-mediated antibody-dependent cell-mediated cytotoxicity in solid tumors: biological evidence and clinical perspectives. Ann Transl Med. 2019;7(5):105.

[19] Zahavi D, AlDeghaither D, O’Connell A, Weiner LM. Enhancing antibody-dependent cell-mediated cytotoxicity: a strategy for improving antibody-based immunotherapy. Antib Ther. 2018;1(1):7-12.

[20] Bournazos S, Wang TT, Dahan R, Maamary J, Ravetch JV. Signaling by Antibodies: Recent Progress. Ann Rev Immunol. 2017;35(1):285-311.

[21] Dixon KJ, Wu J, Walcheck B. Engineering Anti-Tumor Monoclonal Antibodies and Fc Receptors to Enhance ADCC by Human NK Cells. Cancers. 202;13(2):312.

[22] Marshall MJE, Stopforth RJ, Cragg MS. Therapeutic Antibodies: What Have We Learnt from Targeting CD20 and Where Are We Going? Front Immunol. 2017;8:1245.

[23] Payandeh Z, Bahrami AA, Hoseinpoor R, Mortazavi Y, Rajabibazl M, Rahimpour A, et al. The applications of anti-CD20 antibodies to treat various B cell disorders. Biomed Pharmacother. 2019;109:2415-26.

[24] Frisch ES, Pretzsch R, Weber MS. A Milestone in Multiple Sclerosis Therapy: Monoclonal Antibodies Against CD20—Yet Progress Continues. Neurotherapeutics. 2021;18(3):1602-22.

[25] Du FH, Mills EA, Mao-Draayer Y. Next-generation anti-CD20 monoclonal antibodies in autoimmune disease treatment. Autoimmun Highlights. 2017;8(1):1-2.

[26] Roach CA, Cross AH. Anti-CD20 B Cell Treatment for Relapsing Multiple Sclerosis. Front Neurol. 2021;11:595547.

[27] Lee DSW, Rojas OL, Gommerman JL. B cell depletion therapies in autoimmune disease: advances and mechanistic insights. Nat Rev Drug Discov. 2020;20(3):179-99.

[28] McKenna A, Lin GA, Whittington MD, Nikitin D, Herron-Smith S, Campbell JD, et al. Oral and monoclonal antibody treatments for relapsing forms of multiple sclerosis: Effectiveness and value. J Manag Care Spec Pharm. 2023;29(7):857-61.

[29]  Florou D, Katsara M, Feehan J, Dardiotis E, Apostolopoulos V. Anti-CD20 agents for multiple sclerosis: spotlight on ocrelizumab and ofatumumab. Brain Sci. 2020;10(10):758.

[30] Berger JR, Brandstadter R, Bar-Or A. COVID-19, and MS disease-modifying therapies. Neurol Neuroimmunol Neuroinflammation. 2020;7(4).

[31]  Lenz HJ. Management and preparedness for infusion and hypersensitivity reactions. Oncol. 2007;12(5):601-9.

[32] Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, et al. Management of Immune-Related Adverse Events in Patients Treated with Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36(17):1714-68.

[33] Asha MZ, Al-Asaad Y, Khalil SF. The comparative efficacy and safety of anti-CD20 monoclonal antibodies for relapsing-remitting multiple sclerosis: A network meta-analysis. IBRO Neurosci Rep. 2021;11:103-11.

[34] Shah K, Cragg M, Leandro M, Reddy V. Anti-CD20 monoclonal antibodies in systemic lupus erythematosus. Biologicals. 2021;69:1-4.

[35] Weiner GJ. Rituximab: Mechanism of Action. Semin Hematol. 2010;47(2):115-23.

[36] Montalban X, Hauser SL, Kappos L, Arnold DL, Bar-Or A, Comi G, et al. Ocrelizumab versus Placebo in Primary Progressive Multiple Sclerosis. N Engl J Med. 2017;376(3):209-20.

[37] Zhang B. Ofatumumab. MAbs. 2009;1(4):326-31.

[38] Steinman L, Fox E, Hartung HP, Alvarez E, Qian P, Wray S, et al. Ublituximab versus Teriflunomide in Relapsing Multiple Sclerosis. N Engl J Med. 2022;387(8):704-14.

[39]  Ancau M, Berthele A, Hemmer B. CD20 monoclonal antibodies for the treatment of multiple sclerosis: up-to-date. Expert Opin Biol Ther. 2019;19(8):829-43.

[40] Glennie MJ, French RR, Cragg MS, Taylor RP. Mechanisms of killing by anti-CD20 monoclonal antibodies. Mol Immunol. 2007;44(16):3823-37.

[41] Ly S, Nedosekin D, Wong HK. Review of an Anti-CD20 Monoclonal Antibody for the Treatment of Autoimmune Diseases of the Skin. Am J Clin Dermatol. 2023;24(2):247-3.

[42] Mathur S, Sutton J. Personalized medicine could transform healthcare. Biomed Rep. 2017;7(1):3-5.

[43] Costa GD, Comi G. A safety review of current monoclonal antibodies used to treat multiple sclerosis. Expert Opin Drug Saf. 2023.

[44] Sabatino JJ, Zamvil SS, Hauser SL. B-Cell Therapies in Multiple Sclerosis. Cold Spring Harb Perspect Med. 2018;9(2):a032037.

[45] Cencioni MT, Mattoscio M, Magliozzi R, Bar-Or A, Muraro PA. B cells in multiple sclerosis — from targeted depletion to immune reconstitution therapies. Nat Rev Neurol. 2021;17(7):399-414.

[46] Rawal S, Patel MM. Threatening cancer with nanoparticle-aided combination oncotherapy. J Control Release. 2019;301:76-109.

[47] Chua CYX, Jiang AY, Eufrásio-da-Silva T, Dolatshahi-Pirouz A, Langer R, Orive G, et al. Emerging immunomodulatory strategies for cell therapeutics. Trends Biotechnol. 2023;41(3):358-73.

[48] Hernández-Rivas J-Á, Ríos-Tamayo R, Encinas C, Alonso R, Lahuerta JJ. The changing landscape of relapsed and/or refractory multiple myeloma (MM): fundamentals and controversies. Biomark Res. 2022;10(1):1.

[49] Aldeghaither DS, Zahavi DJ, Murray JC, Fertig EJ, Graham GT, Zhang YW, et al. A Mechanism of Resistance to Antibody-Targeted Immune Attack. Cancer Immunol Res. 2019;7(2):230-43.

[50] Frédérich M, Pirotte B, Fillet M, de Tullio P. Metabolomics as a Challenging Approach for Medicinal Chemistry and Personalized Medicine. J Med Chem. 2016;59(19):8649-66.


 

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

International Journal of Pharmaceutical and Phytopharmacological Research
© 2024 All rights reserved