scholarly journals A case for improving frail patient outcomes in multiple myeloma with phenotype‐driven personalized medicine

2021 ◽  
Author(s):  
Lauren T. Reiman ◽  
Zachary J. Walker ◽  
Lyndsey R. Babcock ◽  
Peter A. Forsberg ◽  
Tomer M. Mark ◽  
...  
Author(s):  
Fredrik Schjesvold ◽  
Albert Oriol

A large number of novel treatments for myeloma have been developed and approved, however alkylating drugs continue to be part of standard regimens, additionally, novel alkylators are currently being developed. We performed a non-systematized literary search for relevant papers and communications at large conferences, as well as exploiting the authors knowledge of the field to review the history, current use and novel concepts around traditional alkylators cyclophosphamide, bendamustine and melphalan and current data on the newly developed pro-drug melflufen. Even in the era of targeted treatment and personalized medicine, alkylating drugs continue to be part of standard-of-care in myeloma, and new alkylators are coming to the market.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 532-532 ◽  
Author(s):  
Jonathan J Keats ◽  
David W Craig ◽  
Winnie Liang ◽  
Yellapantula Venkata ◽  
Ahmet Kurdoglu ◽  
...  

Abstract The Multiple Myeloma Research Foundation (MMRF) CoMMpass trial is the cornerstone of the MMRF Personalized Medicine Initiative. The accrual goal is 1000 patients with newly-diagnosed active multiple with sufficient tumor material for the comprehensive analysis of each tumor genome. Each eligible patient will be followed from initial diagnosis longitudinally for a minimum of 8 years. Additional tumor samples will be collected and comprehensively analyzed when possible for each patient at time of suspect CR, recurrence or progression of disease. The clinical study (NCT0145429) opened in July 2011 and now includes 56 sites in the US and Canada that have enrolled over 300 patients as of Aug. 1, 2013. The frontline treatments permitted in this study include current standard of care therapies containing a proteasome inhibitor, an IMiD or both. The comprehensive analysis of each tumor and matched normal genome involves; Long-Insert Whole Genome Sequencing (WGS) to identify somatic copy number alterations and structural changes, Whole Exome Sequencing (WES) to identify somatic single nucleotide variants and indels, and RNA sequencing (RNAseq) to define transcript expression levels and fusion transcripts. In addition, BRAF pyrosequencing and immunophenotyping analysis are being done in CAP-CLIA certified labs. An extensive, open-access, public clinical and molecular database, the CoMMpass Researcher Gateway (RG) (https://research.themmrf.org), is being developed to facilitate the rapid dissemination of the results and provides the myeloma community with a mechanism to analyze the results. The clinical endpoints and outcomes also include Quality of Life measures and health care resource utilization. An initial interim analysis on the first 178 cases has just been completed and made publicly available through the CoMMpass RG. At the molecular level, BRAF analysis on this serial sample set of newly diagnosed patients identified V600E mutations at rate of 5.7%, confirming our previous observations from a mixture of non-consecutive treated and untreated patient samples in our previous genomic efforts. The flow cytometry panel was designed to provide a comprehensive immunofingerprint of each patient that could be used for minimal residual disease monitoring and to monitor potentially therapeutic options; MS4A1/CD20, CD52, KIT/CD117, and FGFR3. These studies have identified tumors which are 100% positive for these actionable antigens at frequencies of; 16.0% for CD20, 5.7% for CD52, 49.7% for CD117, and 8.5% for FGFR3. Of the 178 cases, 34 were profiled through WGS, WES and RNAseq before this interim analysis. We identified 553 variants (median 19 per patient, range 11-55) were the variant allele detected by WES was also detected by RNAseq, suggesting the variant is potentially biologically relevant. Of these genes, 36 were seen more than once and 7 were identified in three or more patients. This includes NRAS (23.5%), KRAS (14.7%), BRAF (8.8%), DIS3 (8.8%), FAM46C (8.8%), TRAF3 (8.8%), and ZNF100 (8.8%). Interestingly, all three ZNF100 variants show preferential expression of the mutant allele. Within this cohort the only recurrent fusion gene identified is the classic IgH-MMSET fusion transcripts associate with t(4;14). The MMRF CoMMpass is providing unprecedented molecular characterization and correlating clinical datasets that will help define the determinants of response to anti-myeloma agents, reveal new, actionable targets and/or those shared with other cancers and facilitate future clinical trial designs, thus serving as a stepping stone toward personalized medicine for myeloma patients. Disclosures: Auclair: MMRC: Employment. Harrison:MMRC: Employment. Jagannath:Celgene: Honoraria; Millennium: Honoraria. Siegel:Celgene: Honoraria, Speakers Bureau; Millennium: Honoraria, Speakers Bureau; Onyx: Honoraria, Speakers Bureau. Vij:Celgene: Honoraria, Research Funding, Speakers Bureau; Millennium: Honoraria, Speakers Bureau; Onyx: Honoraria, Research Funding, Speakers Bureau. Zimmerman:Celgene: Honoraria; Millenium: Honoraria; Onyx: Honoraria. Capone:MMRC: Employment. Lonial:Millennium: Consultancy; Celgene: Consultancy; Novartis: Consultancy; BMS: Consultancy; Sanofi: Consultancy; Onyx: Consultancy.


2016 ◽  
Vol 175 (2) ◽  
pp. 252-264 ◽  
Author(s):  
Kwee Yong ◽  
Michel Delforge ◽  
Christoph Driessen ◽  
Leah Fink ◽  
Alain Flinois ◽  
...  

2021 ◽  
pp. 400-407
Author(s):  
Mercy A. Oduor ◽  
Teresa C. Lotodo ◽  
Terry A. Vik ◽  
Kelvin M. Manyega ◽  
Patrick Loehrer ◽  
...  

Despite improved treatment strategies for multiple myeloma (MM), patient outcomes in low- and middle-income countries remain poor, unlike high-income countries. Scarcity of specialized human resources and diagnostic, treatment, and survivorship infrastructure are some of the barriers that patients with MM, clinicians, and policymakers have to overcome in the former setting. To improve outcomes of patients with MM in Western Kenya, the Academic Model Providing Access to Healthcare (AMPATH) MM Program was set up in 2012. In this article, the program's activities, challenges, and future plans are described distilling important lessons that can be replicated in similar settings. Through the program, training on diagnosis and treatment of MM was offered to healthcare professionals from 35 peripheral health facilities across Western Kenya in 2018 and 2019. Access to antimyeloma drugs including novel agents was secured, and pharmacovigilance systems were developed. Finally, patients were supported to obtain health insurance in addition to receiving peer support through participation in support group meetings. This article provides an implementation blueprint for similar initiatives aimed at increasing access to care for patients with MM in underserved areas.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3000-3000
Author(s):  
Maria Teresa Petrucci ◽  
João Mendes ◽  
Jennifer H. Boer ◽  
Gabriele Casamassima ◽  
Anna Willis ◽  
...  

Abstract Introduction: Multiple myeloma (MM) is an incurable disease characterized by the proliferation of malignant plasma cells within the bone marrow, causing a wide range of burdensome symptoms. Patients initiating treatment typically receive a combination of drugs across various classes with or without autologous stem cell transplantation (ASCT). However, patients will invariably relapse following initial treatment, and often require many lines of drug treatment over the course of their disease. Real-word data showed that a significant proportion of newly diagnosed MM patients that receive frontline (FL) treatment did not receive subsequent treatment. These high attrition rates suggest that using the best treatment upfront is crucial in delaying disease progression. The CASSIOPEIA (transplant-eligible [TE] setting), MAIA and ALCYONE (transplant-ineligible [TIE] setting) trials demonstrate that the addition of daratumumab (DARA) to standard of care treatments in FL significantly improves patient outcomes. Based on data from these trials, the European marketing authorization for DARA has been extended to the FL setting. To ensure the best possible long-term patient outcomes in clinical practice, the availability of new FL treatment options requires a redefinition of treatment patterns. Thus, we aim to investigate whether the adoption of DARA as a FL, as opposed to later-line, treatment of MM leads to better outcomes and improved clinical practice. Methods: In the absence of real-world sequencing data, we developed a clinical sequencing simulation using individual patient data from the DARA trials and indirect comparative evidence, across all indications in MM. We used progression-free survival curves to simulate health state transition probabilities across four lines of active treatment, to capture the efficacy of treatment sequences in MM. Patients start with initiation of FL treatment, and ASCT eligibility determines the sequences patients receive. Clinical expert opinion was sought to determine 1) the full range of meaningful treatment sequences and 2) which of these are used most in Italian clinical practice. Based on the clinical simulation outcomes, we calculated average time spent in each line of treatment, percentage of patients alive at different timepoints, and the total survival for patients initiating a sequence. This analysis included conservative attrition rates from trial data, 14% for TE (CASSIOPEIA) and 24% for TIE (MAIA/ALCYONE), assumed as similar across regimens in each setting. Results: In the TE setting, the best outcomes were achieved when using the DARA-based regimen (DVTd) as FL treatment, followed by either a LEN-based regimen (KRd) or a BOR-based regimen (PVd), resulting in a total survival of 14.2 and 14.1 years, respectively. In the TIE setting, the best outcomes were achieved when DRd or DVMP were used as FL treatment, followed by either a BOR-based regimen (PVd, for DRd) or a LEN-based regimen (KRd, for DVMP), resulting in a total survival of 11.7 and 10.9 years, respectively. In both the FL and second line (2L) settings, there was a clear survival benefit of using DARA. When comparing the DARA-based sequence with the current FL TE benchmark sequence (DVTd + KRd + Pd + Vd versus VTd + DRd + Kd + Pd), an additional survival of 1.5 years was observed in TE patients. When DARA was added to the current FL TIE benchmark sequence (DRd + PVd + Kd + Vd versus VMP + DRd + Kd + Pd), TIE patients lived on average 2.8 years longer. For TE patients, time spent progression-free ranged from an average of 4.83 to 7.99 years at FL, 1.42 to 5.40 years in 2L, 0.23 to 2.24 years in 3L and 0.17 to 1.53 years on 4L. For TIE patients, the variation was higher, leaving more room for optimization: 1.97 to 7.31 years at FL, 0.68 to 4.76 years in 2L, 0.17 to 3.25 years in 3L and 0.19 to 0.51 years in 4L. Conclusion: To our knowledge, this is the first sequencing simulation to consider optimal patient outcomes across several lines of MM treatment. The results show that the longest time in remission is achieved with the use of DARA-based regimens as FL treatment, significantly improving patient outcomes. Time spent progression free decreases with each subsequent line of treatment and the magnitude of the effect seen in the third and fourth treatment lines is not as significant as that of the effect seen in earlier treatment lines. Therefore, patients should be treated with the most effective treatment upfront. Disclosures Petrucci: Celgene: Honoraria, Other: Advisory Board; Janssen-Cilag: Honoraria, Other: Advisory Board; BMS: Honoraria, Other: Advisory Board; Takeda: Honoraria, Other: Advisory Board; Amgen: Honoraria, Other: Advisory Board; GSK: Honoraria, Other: Advisory Board; Karyopharm: Honoraria, Other: Advisory Board. Mendes: Janssen-Cilag Farmacêutica: Current Employment. Boer: Janssen: Consultancy. Casamassima: Janssen: Current Employment. Willis: Janssen: Consultancy. Wadlund: Janssen: Current Employment. Matthijsse: Janssen: Consultancy. Armeni: Astrazeneca: Consultancy; Boehringer Ingelheim: Consultancy; Novartis: Consultancy; Sanofi: Consultancy; Johnson & Johnson: Consultancy; Amgen: Consultancy; Janssen: Consultancy.


2016 ◽  
Vol 34 (3_suppl) ◽  
pp. e270-e270
Author(s):  
Sherri Z. Millis ◽  
John M Davis ◽  
Stephanie Marie Ratliff ◽  
Melissa Lorraine Ray ◽  
Wendy M Schroeder ◽  
...  

e270 Background: Cancer treatment based on an individual’s tumor profiling has been associated with increased time to progression. Despite this, adoption has been impeded by various technical, financial, legislative, and ethical issues. Implementation of a personalized medicine program into a healthcare system with a goal of improving patient outcomes includes clinician and patient education, increased tumor profiling of patients, data sharing/analysis, expanded research, and billing/reimbursement practices. Methods: A personalized medicine program was implemented at a hospital system. Measures of physician practice included attendance at genomic medicine education sessions, utilization of genetic/genomic tests, enrollment of patients into clinical trials, modifications in billing practices, and submission of data for analysis. Reimbursement of off-label pharmaceutical agents as well as use of aggregate data to inform treatment and enrollment in biomarker-based clinical trials was recorded. Frequency of genetic and/or genomic tests relative to physician and patient education and access to research opportunities was also assessed. Early patient outcomes, overall costs of care, access to clinical trials, and changes in knowledge and communication are also being monitored. Results: Although analysis is ongoing, the initial assessment indicates an increased utilization of genetic and genomic tests, clarity in billing practices, improved reimbursement for off label therapies, and consumption of educational opportunities from clinicians to patients/caregivers. Analysis of the first year implementation will be reported, including evaluation of preliminary results relative to quality of life and survivorship. Conclusions: Preliminary analysis of data from implementation of a personalized medicine program indicates that utilizing relevant education, research, aggressive billing and reimbursement processes, and IT infrastructure, can provide patients with the individual therapies which reduce cost and improve survivorship.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1818-1818
Author(s):  
Lin Yang ◽  
Xingding Zhang ◽  
Hua Wang ◽  
Robert Z. Orlowski ◽  
Xingxu Huang

Abstract Abstract 1818 Chemotherapy agents are extremely important in the treatment of liquid malignancies, such as multiple myeloma (MM). Unfortunately, chemotherapy resistance in MM therapy is the most significant cause of treatment failure. The ability to predict, treat, or circumvent resistance is extremely likely to improve clinical outcomes. Thus, identification of novel genes that play a crucial role in MM progression and chemosensitivity is necessary to understand this disease better at the molecular level. Moreover, these genes and their products may serve as new therapeutic targets for MM, whose expression could improve patient outcomes or served as a predictor for chemotherapy outcome. To identify potential chemosensitivity genes, establishing a high-throughput method for validation of targets becomes urgently needed. Toward this purpose, we have successfully developed a high-throughput siRNA based functional target validation approach and identified 34 potential chemosensitivity genes. Our preliminary studies focusing on one of the candidate gene, TJP1 (tight junction protein 1), suggested that targeting TJP1 led to tumor cell resistant to several chemotherapy agents, including doxorubicin (Dox), cisplatin (Cis), methotrexate (MTX), and bortezomib. Further analysis with 264 bortezomib treated MM patients indicated that expression level of TJP1 correlated with patient response to bortezomib. Two clones and pooled RPMI 8226 MM cell line, which were developed against bortezomib treatment in our lab, showed loss of TJP1 expression, suggesting a role of TJP1 may play in bortezomib resistance development. More importantly, TJP1 targeting in myeloma cells resulted in cell resistance to bortezomib treatment. Protein tyrosine phosphatase, receptor type, O1 (PTRPO) was identified in our screening as well. We first examined the expression of PTPRO in a panel of myeloma cell lines, and found that PTPRO expression was dramatically decreased in several drug resistant lines compared to their parental sensitive cells. Moreover, using a Lentiviral cDNA expression system to overexpress PTPRO, we observed that PTPRO significantly inhibited cell growth and sensitized drug sensitivity of several drug lines by triggering apoptosis. Finally, evaluation of the publicly available gene expression profiling data gathered from primary plasma cells on the Multiple Myeloma Genomics Portal, and the linked clinical annotation describing patient outcomes, has shown that over-expression of PTPRO is associated with a superior overall survival compared to patients whose myeloma show low levels of PTRPO expression, suggesting this may be a prognostic marker as well. Together, we have successfully identified a group of potential chenmosensitivity genes, in which two of the genes (TJP1 and PTPRO) have been further validated to sensitize the drug treatment in multiple myeloma cells. Disclosures: No relevant conflicts of interest to declare.


Cells ◽  
2021 ◽  
Vol 10 (2) ◽  
pp. 336
Author(s):  
Ignacio J. Cardona-Benavides ◽  
Cristina de Ramón ◽  
Norma C. Gutiérrez

Some genetic abnormalities of multiple myeloma (MM) detected more than two decades ago remain major prognostic factors. In recent years, the introduction of cutting-edge genomic methodologies has enabled the extensive deciphering of genomic events in MM. Although none of the alterations newly discovered have significantly improved the stratification of the outcome of patients with MM, some of them, point mutations in particular, are promising targets for the development of personalized medicine. This review summarizes the main genetic abnormalities described in MM together with their prognostic impact, and the therapeutic approaches potentially aimed at abrogating the undesirable pathogenic effect of each alteration.


2021 ◽  
pp. 107815522110152
Author(s):  
Noopur Raje ◽  
Rohan Medhekar ◽  
Sumeet Panjabi ◽  
Dionne M Hines ◽  
Xin Wang ◽  
...  

Introduction Carfilzomib dosing as a single agent or in combination with dexamethasone (Kd) has evolved from the initial 27 mg/m2 twice-weekly (legacy dose), to more recently approved doses of 56 mg/m2 twice-weekly and 70 mg/m2 once-weekly (optimized doses). The objective of this study was to evaluate the overall survival (OS), and time to next treatment (TTNT) among multiple myeloma patients treated with Kd optimized vs legacy doses. Methods A retrospective analysis of patients receiving Kd between 01/01/2013–07/31/2017 was conducted using IQVIA’s oncology electronic medical records database. Kd dose was estimated based on body surface area. OS was measured from the Kd-initiation date until death. TTNT was defined as the time from Kd-initiation until the start of subsequent treatment. Kaplan-Meier analysis and Cox models were used to evaluate OS and TTNT. Results Of the 1,469 patients evaluated, 129 (8.8%) received optimized dose and 1,340 (91.2%) received legacy dose. Risk of mortality was 64% lower for patients receiving the optimized doses (HR: 0.36, 95% CI: 0.178–0.745). Patients receiving the optimized doses had significantly longer TTNT compared to patients receiving the legacy dose (median TTNT: 17.5 months [95% CI: 14.8-NE] and 13.2 months, [95% CI: 12.4–14.4], respectively; p = 0.023), and 33% lower risk of progressing to the subsequent treatment (HR: 0.67, 95% CI: 0.48–0.93). Conclusions Patient outcomes could be improved if eligible MM patients are treated with the optimized, recently approved Kd doses (56 mg/m2 twice-weekly and 70 mg/m2 once-weekly).


Hematology ◽  
2013 ◽  
Vol 2013 (1) ◽  
pp. 478-487 ◽  
Author(s):  
Ola Landgren

Abstract After decades of virtually no progress, multiple myeloma survival has improved significantly in the past 10 years. Indeed, multiple myeloma has perhaps seen more remarkable progress in treatment and patient outcomes than any other cancer during the last decade. Recent data show that multiple myeloma is consistently preceded by a precursor state (monoclonal gammopathy of undetermined significance [MGUS]/smoldering multiple myeloma [SMM]). This observation provides a framework for prospective studies focusing on transformation from precursor disease to multiple myeloma and for the development of treatment strategies targeting “early myeloma.” This review discusses current biological insights in MGUS/SMM, provides an update on clinical management, and discusses how the integration of novel biological markers, molecular imaging, and clinical monitoring of MGUS/SMM could facilitate the development of early treatment strategies for high-risk SMM (early myeloma) patients in the future.


Sign in / Sign up

Export Citation Format

Share Document