(Peer-reviewed, Open Access, Fast processing International Journal) Impact Factor : 5.0 , ISSN 0525-1003
Offer for Students ₹ 999 INR ( offer valid till 31st December 2025)
(Peer-reviewed, Open Access, Fast processing International Journal) Impact Factor : 5.0 , ISSN 0525-1003
Volume 06, Issue 12.17 , December , 2025
17. Multiple Myeloma: Pathogenesis, Clinical Features, Diagnostic Advances, and Evolving Therapeutic Strategies
1. Dr Turdaliev Samatbek [https://orcid.org/0000-0002-0836-0671]
2. Manal Najeeb
( 1, Teacher, International Medical Faculty, Osh State University; Republic of Kyrgyzstan
2, Student Group 15, 5th Year, International Medical Faculty, Osh State University; Republic of Kyrgyzstan )
Abstract
Multiple myeloma( MM) is a clonal plasma cell malignancy characterized by bone marrow infiltration, monoclonal protein production, and end- organ damage. It accounts for roughly 10 of hematologic malice and remains incurable despite significant remedial advances. Over the last two decades, improved understanding of disease biology, cytogenetics, and the bone marrow medium has transformed diagnosis and management. new agents similar as proteasome inhibitors, immunomodulatory medicines, monoclonal antibodies, and cellular therapies have markedly bettered survival. This composition reviews the current understanding of the pathophysiology, clinical presentation, individual criteria, threat position, and treatment modalities of multiple myeloma, pressing recent advances and ongoing challenges.
Introduction
Multiple myeloma is a malignant plasma cell dyscrasia arising frompost-germinal center B lymphocytes that have undergone physical hypermutation and immunoglobulin class- switch recombination, resulting in the clonal proliferation of antibody- secreting plasma cells( Rajkumar & Kumar, 2020). The complaint generally evolves through well- honored premalignant and asymptomatic stages, most specially monoclonal gammopathy of undetermined significance( MGUS) and smoldering multiple myeloma, both of which are characterized by the presence of monoclonal immunoglobulin without overt end- organ damage( Kyle et al., 2018). Longitudinal studies have demonstrated that MGUS progresses to multiple myeloma or related plasma cell disorders at an approximate rate of 1 per year, highlighting the significance of early identification and surveillance.
Multiple myeloma generally affects aged adults, with a median age at opinion ranging between 65 and 70 years, and exhibits a clear manly predominance( Siegel et al., 2024). Epidemiological data further indicate advanced incidence rates among certain ethnical populations and individualities with a family history of plasma cell disorders, suggesting a combination of inheritable susceptibility and environmental influences in disease development. Advancing age contributes significantly to complaint threat, probably due to accretive inheritable mutations and age- related vulnerable dysregulation.
Clinically, multiple myeloma is defined by the infiltration of clonal plasma cells within the bone marrow, leading to the production of monoclonal immunoglobulin( M- protein) and posterior organ dysfunction. The hallmark clinical manifestations are classically epitomized by the CRAB criteria, which include hypercalcemia resulting from increased bone resorption, renal impairment due to light- chain deposit and tubular injury, anemia secondary to bone gist repression, and lytic bone lesions caused by enhanced osteoclastic exertion and suppressed osteoblastic function( Dimopoulos et al., 2015). Cases constantly present with bone pain, fatigue, intermittent infections, and renal dysfunction, significantly affecting quality of life.
Although multiple myeloma remains an incurable malignancy, patient issues have bettered mainly over the once two decades. The preface of new remedial agents, including proteasome impediments, immunomodulatory medicines, monoclonal antibodies, and cellular curatives, combined with threat- acclimated and personalized treatment strategies, has redounded in pronounced advancements in progression-free and overall survival. Advances in individual criteria, early complaint discovery, and probative care have further contributed to enhanced complaint control and dragged survival, transubstantiating multiple myeloma into a habitual, manageable condition for numerous cases.
Materials and Methods
This article is a narrative review based on an extensive and methodical literature search conducted using major biomedical databases, including PubMed and Google Scholar, along with internationally recognized hematology and oncology guidelines issued by authoritative bodies. Peer- reviewed original research articles, randomized controlled trials, methodical reviews, meta- analyses, and expert consensus guidelines published generally between 2010 and 2024 were considered eligible for addition. The hunt strategy employed a combination of Medical Subject Headings( MeSH) terms and free- text keywords, including multiple myeloma, plasma cell dyscrasia, proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, new targeted therapies, and autologous stem cell transplantation. fresh search filters were applied to prioritize mortal studies and publications in the English language. To ensure comprehensive coverage and minimize the risk of omitting applicable data, the reference lists of selected high- impact papers and guideline documents were manually reviewed. Emphasis was placed on studies that significantly contributed to the understanding of complaint pathogenesis, individual criteria, threat position, and evolving remedial strategies in multiple myeloma. The collected literature was critically rated and synthesized to give an over- to- date and clinically applicable overview of the complaint.
Results
Pathogenesis and Disease Biology
Multiple myeloma arises from a multistep process involving inheritable abnormalities and interactions with the bone marrow microenvironment. Primary inheritable events include immunoglobulin heavy- chain( IgH) translocations similar as t( 11; 14), t( 4; 14), and t( 14; 16), or hyperdiploidy( Fonseca et al., 2009). Secondary events include mutations in KRAS, NRAS, TP53, and MYC dysregulation( Walker et al., 2015).
Myeloma cells interact with stromal cells via cytokines similar as interleukin- 6( IL- 6), vascular endothelial growth factor( VEGF), and receptor activator of nuclear factor κB ligand( RANKL), leading to osteoclast activation and osteoblast suppression, explaining the characteristic lytic bone disease( Roodman, 2010).
Clinical Features
Patients generally present with
• Bone pain( especially spine and ribs)
• Fatigue due to anemia
• intermittent infections
• Renal impairment
• Hypercalcemia- related symptoms
Extramedullary complaint and tube cell leukemia represent aggressive variants associated with poor prognosis( Usmani et al., 2012).
Diagnosis is grounded on International Myeloma Working Group( IMWG) criteria, which incorporate biomarkers enabling earlier diagnosis before unrecoverable organ damage( Rajkumar et al., 2014).
Table 1. Major Drug Classes Used in Multiple Myeloma
Autologous stem cell transplantation (ASCT) remains standard for eligible patients and improves progression-free survival (Attal et al., 2017). Novel therapies such as CAR-T cells targeting BCMA and bispecific antibodies show promising results in relapsed/refractory disease (Munshi et al., 2021).
Discussion
The management of multiple myeloma has evolved from conventional chemotherapy to biologically targeted therapies. Early diagnosis using biomarker-driven criteria allows timely intervention. However, challenges remain, including clonal heterogeneity, drug resistance, and disease relapse. Resource limitations in low- and middle-income countries further complicate management, emphasizing the need for cost-effective strategies and early screening.
Future directions include personalized therapy guided by molecular profiling, minimal residual disease (MRD) monitoring, and expanded use of immunotherapies.
Conclusion
Multiple myeloma is a complex and heterogeneous hematologic malignancy associated with considerable morbidity and mortality, largely due to its multisystem involvement and tendency toward disease relapse. Over recent decades, significant advances in the understanding of disease biology, including genetic heterogeneity, bone marrow microenvironment interactions, and mechanisms of treatment resistance, have fundamentally transformed the clinical management of the disease. The introduction of novel therapeutic agents such as proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, and emerging cellular therapies has resulted in substantial improvements in response rates, progression-free survival, and overall survival across diverse patient populations.
Despite these achievements, multiple myeloma remains incurable, and long-term disease control continues to pose a major clinical challenge. Ongoing research efforts focusing on early disease detection, refined risk stratification, and individualized, risk-adapted treatment strategies are essential to optimize outcomes. In addition, the development of durable immunologic control through advanced immunotherapies, minimal residual disease guided treatment approaches, and combination regimens holds promise for achieving deeper and more sustained remissions. Continued translational and clinical research, coupled with equitable access to modern therapies, will be critical in further improving survival, reducing disease-related complications, and enhancing the overall quality of life for patients with multiple myeloma.
References
Rajkumar SV, Kumar S. Multiple myeloma: diagnosis and treatment. Mayo Clin Proc. 2020.
Kyle RA et al. Monoclonal gammopathy of undetermined significance. N Engl J Med. 2018.
Siegel RL et al. Cancer statistics, 2024. CA Cancer J Clin. 2024.
Dimopoulos MA et al. IMWG updated criteria. Lancet Oncol. 2015.
Fonseca R et al. Genetic heterogeneity in myeloma. Blood. 2009.
Walker BA et al. Mutational spectrum in myeloma. Nature. 2015.
Roodman GD. Pathogenesis of myeloma bone disease. Leukemia. 2010.
Usmani SZ et al. Extramedullary myeloma. Leukemia. 2012.
Rajkumar SV et al. Biomarker-defined MM. Lancet Oncol. 2014.
Palumbo A et al. Revised ISS. J Clin Oncol. 2015.
Attal M et al. ASCT in myeloma. N Engl J Med. 2017.
Munshi NC et al. CAR-T therapy. N Engl J Med. 2021.
Kumar SK et al. Improved survival in MM. Blood. 2014.
Dispenzieri A et al. Light chain amyloidosis. Blood. 2012.
Sonneveld P et al. Bortezomib-based therapy. Blood. 2016.
Richardson PG et al. Proteasome inhibition. Blood. 2018.
Lonial S et al. Daratumumab therapy. N Engl J Med. 2016.
Moreau P et al. Carfilzomib outcomes. Lancet. 2015.
Dimopoulos MA et al. Lenalidomide maintenance. Blood. 2012.
Landgren O et al. Smoldering myeloma. J Clin Oncol. 2020.
Kumar S et al. MRD negativity. Blood. 2016.
Avet-Loiseau H et al. Cytogenetic risk. Blood. 2007.
Anderson KC. Role of microenvironment. Nat Rev Cancer. 2007.
Mateos MV et al. Early treatment in SMM. N Engl J Med. 2013.
San Miguel JF et al. Treatment algorithms. Blood. 2020.
Dimopoulos MA et al. Renal impairment. Leukemia. 2008.
Terpos E et al. Bone disease management. Lancet Oncol. 2018.
Gay F et al. Continuous therapy. Blood. 2015.
Bianchi G, Anderson KC. Immunotherapy in MM. Blood. 2014.
Durie BGM et al. Myeloma management guidelines. Blood Cancer J. 2017.