scholarly journals Measurement of ex vivo resistance to proteasome inhibitors, IMiDs, and daratumumab during multiple myeloma progression

2020 ◽  
Vol 4 (8) ◽  
pp. 1628-1639
Author(s):  
Zachary J. Walker ◽  
Michael J. VanWyngarden ◽  
Brett M. Stevens ◽  
Diana Abbott ◽  
Andrew Hammes ◽  
...  

Abstract The oncogenic drivers and progression factors in multiple myeloma (MM) are heterogeneous and difficult to target therapeutically. Many different MM drugs have emerged, however, that attack various phenotypic aspects of malignant plasma cells. These drugs are administered in numerous, seemingly interchangeable combinations. Although the availability of many treatment options is useful, no clinical test capable of optimizing and sequencing the treatment regimens for an individual patient is currently available. To overcome this problem, we developed a functional ex vivo approach to measure patients’ inherent and acquired drug resistance. This method, which we termed myeloma drug sensitivity testing (My-DST), uses unselected bone marrow mononuclear cells with a panel of drugs in clinical use, followed by flow cytometry to measure myeloma-specific cytotoxicity. We found that using whole bone marrow cultures helped preserve primary MM cell viability. My-DST was used to profile 55 primary samples at diagnosis or at relapse. Sensitivity or resistance to each drug was determined from the change in MM viability relative to untreated control samples. My-DST identified progressive loss of sensitivity to immunomodulatory drugs, proteasome inhibitors, and daratumumab through the disease course, mirroring the clinical development of resistance. Prospectively, patients’ ex vivo drug sensitivity to the drugs subsequently received was sensitive and specific for clinical response. In addition, treatment with <2 drugs identified as sensitive by My-DST led to inferior depth and duration of clinical response. In summary, ex vivo drug sensitivity is prognostically impactful and, with further validation, may facilitate more personalized and effective therapeutic regimens.

2021 ◽  
pp. 602-612
Author(s):  
David G. Coffey ◽  
Andrew J. Cowan ◽  
Bret DeGraaff ◽  
Timothy J. Martins ◽  
Niall Curley ◽  
...  

PURPOSE Multiple myeloma (MM) is a genetically heterogeneous malignancy characterized by variable treatment responses. Although numerous drugs have been approved in recent years, the ability to predict treatment response and tailor individual therapy is limited by the absence of robust predictive biomarkers. The goal of this clinical trial was to use ex vivo, high-throughput screening (HTS) of 170 compounds to predict response among patients with relapsed or refractory MM and inform the next treatment decisions. Additionally, we integrated HTS with multi-omic analysis to uncover novel associations between in vitro drug sensitivity and gene expression and mutation profiles. MATERIALS AND METHODS Twenty-five patients with relapsed or refractory MM underwent a screening bone marrow or soft tissue biopsy. Sixteen patients were found to have sufficient plasma cells for HTS. Targeted next-generation sequencing was performed on plasma cell-free DNA from all patients who underwent HTS. RNA and whole-exome sequencing of bone marrow plasma cells were performed on eight and seven patients, respectively. RESULTS Results of HTS testing were made available to treating physicians within a median of 5 days from the biopsy. An actionable treatment result was identified in all 16 patients examined. Among the 13 patients who received assay-guided therapy, 92% achieved stable disease or better. The expression of 105 genes and mutations in 12 genes correlated with in vitro cytotoxicity. CONCLUSION In patients with relapsed or refractory MM, we demonstrate the feasibility of ex vivo drug sensitivity testing on isolated plasma cells from patient bone marrow biopsies or extramedullary plasmacytomas to inform the next line of therapy.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3876-3876 ◽  
Author(s):  
Andrzej J Jakubowiak ◽  
William Bensinger ◽  
David Siegel ◽  
Todd M. Zimmerman ◽  
Jan M. Van Tornout ◽  
...  

Abstract Abstract 3876 Poster Board III-812 Background Elotuzumab is a humanized monoclonal IgG1 antibody directed against CS1, a cell surface glycoprotein, which is highly and uniformly expressed in multiple myeloma (MM). In mouse xenograft models of MM, elotuzumab demonstrated significantly enhanced anti-tumor activity when combined with bortezomib compared to bortezomib alone (Van Rhee et al., Mol. Cancer Ther., in press, 2009). This phase 1/2 trial will determine the maximum tolerated dose (MTD), overall safety, pharmacokinetics (PK) and clinical response of elotuzumab in combination with bortezomib in patients with relapsed MM following 1-3 prior therapies. Methods The study consists of 4 escalating cohorts of elotuzumab (2.5 mg/kg to 20 mg/kg) administered on Days 1 and 11 and bortezomib (1.3 mg/m2) administered on Days 1, 4, 8 and 11 of a 21-day cycle. Patients with progressive disease at the end of Cycle 2 or 3 also receive oral dexamethasone (20 mg) on Days 1, 2, 4, 5, 8, 9, 11 and 12 of each subsequent cycle. Patients with stable disease or better at the end of 4 cycles will continue treatment for 6 or more cycles unless withdrawn earlier due to unexpected toxicity or disease progression. Key entry criteria: age ≥ 18 years; confirmed diagnosis of MM and documentation of 1 to 3 prior therapies; measurable disease M-protein component in serum and/or in urine; and no prior bortezomib treatment within 2 weeks of first dose. Results To date, a total of 16 MM patients with a median age of 64 years have been enrolled in the study. The median time from initial diagnosis of MM was 3.5 years and patients had received a median of 2 prior MM treatments. Patients have been treated in four cohorts; 3 each in 2.5, 5 and 10 mg/kg elotuzumab cohorts, and 7 in the 20 mg/kg elotuzumab cohort. No dose limiting toxicity (DLT) was observed during the first cycle of the study and the MTD was not established. Five SAEs have been reported in four patients in later treatment cycles; two events, chest pain and gastroenteritis, occurring in one patient, were considered elotuzumab-related. Other SAEs include grade 3 sepsis, vomiting, pneumonia and grade 2 dehydration. The most common AEs reported include Grade 1-3 diarrhea, constipation, nausea, fatigue, thrombocytopenia, neutropenia, anemia and peripheral neuropathy. The best clinical response (EBMT criteria) for the 16 patients who have received at least two cycles of treatment is shown in the table below. Preliminary PK analysis suggests a serum half-life of 10-11 days at higher doses (10 and 20 mg/kg). Preliminary analysis of peripheral blood mononuclear cells and bone marrow of patients on study indicates that objective responses in the study correlate well with complete saturation of CS1 sites by elotuzumab on bone marrow plasma and NK cells. Conclusions The combination of elotuzumab with bortezomib has a manageable adverse event profile and shows promising preliminary efficacy with ≥PR in 44% and ≥MR in 75% of all enrolled patients. Accrual is ongoing in the expanded 20 mg/kg cohort. Updated safety, efficacy, and PK data will be presented at the meeting. Disclosures: Jakubowiak: Millennium: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Centocor Ortho Biotech: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Exelixis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Off Label Use: Bortezomib in combination with elotuzumab for the treatment of relapsed/refractory multiple myeloma. Bensinger:Millennium: Membership on an entity's Board of Directors or advisory committees. Siegel:Millennium: Speakers Bureau; Celgene: Speakers Bureau. Zimmerman:Millennium: Speakers Bureau; Centecor: Speakers Bureau. Van Tornout:BMS: Employment. Zhao:Facet Biotech: Employment. Singhal:Facet Biotech: Employment. Anderson:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Millennium: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Kinan Alhallak ◽  
Amanda Jeske ◽  
Pilar de la Puente ◽  
Jennifer Sun ◽  
Mark Fiala ◽  
...  

AbstractCancer patients undergo detrimental toxicities and ineffective treatments especially in the relapsed setting, due to failed treatment attempts. The development of a tool that predicts the clinical response of individual patients to therapy is greatly desired. We have developed a novel patient-derived 3D tissue engineered bone marrow (3DTEBM) technology that closely recapitulate the pathophysiological conditions in the bone marrow and allows ex vivo proliferation of tumor cells of hematologic malignancies. In this study, we used the 3DTEBM to predict the clinical response of individual multiple myeloma (MM) patients to different therapeutic regimens. We found that while no correlation was observed between in vitro efficacy in classic 2D culture systems of drugs used for MM with their clinical efficacious concentration, the efficacious concentration in the 3DTEBM were directly correlated. Furthermore, the 3DTEBM model retrospectively predicted the clinical response to different treatment regimens in 89% of the MM patient cohort. These results demonstrated that the 3DTEBM is a feasible platform which can predict MM clinical responses with high accuracy and within a clinically actionable time frame. Utilization of this technology to predict drug efficacy and the likelihood of treatment failure could significantly improve patient care and treatment in many ways, particularly in the relapsed and refractory setting. Future studies are needed to validate the 3DTEBM model as a tool for predicting clinical efficacy.


2015 ◽  
Vol 15 ◽  
pp. e286
Author(s):  
M.M. Majumder ◽  
R. Silvennoinen ◽  
P. Anttila ◽  
D. Tamborero ◽  
S. Eldfors ◽  
...  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ciara Tierney ◽  
Despina Bazou ◽  
Muntasir M. Majumder ◽  
Pekka Anttila ◽  
Raija Silvennoinen ◽  
...  

AbstractWith the introduction of novel therapeutic agents, survival in Multiple Myeloma (MM) has increased in recent years. However, drug-resistant clones inevitably arise and lead to disease progression and death. The current International Myeloma Working Group response criteria are broad and make it difficult to clearly designate resistant and responsive patients thereby hampering proteo-genomic analysis for informative biomarkers for sensitivity. In this proof-of-concept study we addressed these challenges by combining an ex-vivo drug sensitivity testing platform with state-of-the-art proteomics analysis. 35 CD138-purified MM samples were taken from patients with newly diagnosed or relapsed MM and exposed to therapeutic agents from five therapeutic drug classes including Bortezomib, Quizinostat, Lenalidomide, Navitoclax and PF-04691502. Comparative proteomic analysis using liquid chromatography-mass spectrometry objectively determined the most and least sensitive patient groups. Using this approach several proteins of biological significance were identified in each drug class. In three of the five classes focal adhesion-related proteins predicted low sensitivity, suggesting that targeting this pathway could modulate cell adhesion mediated drug resistance. Using Receiver Operating Characteristic curve analysis, strong predictive power for the specificity and sensitivity of these potential biomarkers was identified. This approach has the potential to yield predictive theranostic protein panels that can inform therapeutic decision making.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3203-3203
Author(s):  
Zachary J Walker ◽  
Michael J VanWyngarden ◽  
Brett M Stevens ◽  
Christophe Langouët-Astrie ◽  
Clayton Smith ◽  
...  

Abstract Background: The oncogenic drivers and progression factors in multiple myeloma (MM) are heterogeneous and difficult to target therapeutically. As a result, personalized medicine approaches have not yet been realized. However, clinical availability of numerous anti-myeloma drugs and readily obtainable bone marrow (BM) aspirates raises the possibility to benefit patients by profiling the drug sensitivity of their MM. Despite newly available drugs, resistance inevitably develops and there is no defined rationale for sequencing therapy. Treatments now consist of >15 agents, including IMiDs, protease inhibitors (PIs), alkylator chemotherapies, steroids, monoclonal antibodies, and options are still expanding. To profile sensitivity to the wide array of options, we developed a rapid functional assay of primary MM cells, distinct from prior assays by forgoing CD138+ cell selection to optimize MM cell viability and allow measurement of monoclonal antibody activity. Methods: Myeloma drug sensitivity testing (My-DST) was performed on patient BM aspirates to measure the MM sensitivity in heterogeneous mononuclear cell (MNC) mixtures. My-DST is performed by culturing samples with a 7-drug panel consisting of bortezomib (Bor), carfilzomib (Car), lenalidomide (Len), pomalidomide (Pom), dexamethasone (Dex), cyclophosphamide (Cy, metabolite 4HC), and daratumumab (Dara) for 48h, followed by multiparameter, high-throughput flow cytometry to measure the surviving MM population. All conditions are performed in triplicate and in parallel, clonality verified by kappa/lambda staining. Results: MM cell viability was substantially better in unselected MNC cultures than CD138-selected cultures (data not shown). Dose-response was evaluated for each agent in MNC cultures to select single active concentrations for high throughput screening (data not shown). Single concentration profiling was completed on 55 patient samples from diagnosis (n = 24), first relapse (n = 11) and after multiple relapses (n = 20) with 2.5 nM PIs, 10 µM IMiDs, 1 µM Dex, 3.75 µM Cy and 20 nM Dara. Each was scored as sensitive or resistant using a cutoff of 80% surviving MM cells normalized to untreated controls (Fig 1A). Mild inherent resistance to varied tested agents was evident at diagnosis and progressively increased with lines of therapy (LOT) until multidrug resistance predominated (Fig 1B). My-DST displayed significant correlation for LOT with increasing resistance to Bor (r = 0.27, P = 0.046), Car (r = 0.27, P = 0.044), Len (r = 0.44, P = 0.0006) and Pom (r = 0.43, P = 0.0009), but not Dara (r = 0.27, P = 0.10), Cy (r = 0.17, P = 0.24) and Dex (r = 0.032, P = 0.82) (Fig 1C). The ex vivo sensitivity for Bor and Car were highly correlated (r = 0.67, P < 0.0001) from sensitive to both to resistant to both, but still differential results favoring Bor or Car were observed (Fig 1D). The ex vivo results for the tested IMiDs were even more highly correlated (r = 0.79, P < 0.0001), with fewer samples showing greater sensitivity differentially to Len or Pom (data not shown). In patients at diagnosis, the ex vivo sensitivity to all drugs given during treatment led to significantly deeper clinical responses than those with resistance to ≥1 drugs received (Fig 1E). As an example of potential clinical utility of My-DST, the test for #847 at diagnosis identified sensitivity to PIs and resistance to IMiDs, which correlated with inadequate response to Len/Dex, followed by complete response (CR) after change to Bor/Dex (Fig 1F). Dara had ex vivo activity in most naïve patients, but patients already exposed showed significant acquired resistance (Fig 1G). We also found that CD38 expression was significantly lower in patients that showed ex vivo Dara resistance (Fig 1H). The ex vivo Dara activity was partially blocked by inhibiting macrophages with clodronate-liposomes (CL) (Fig 1I). Conclusion: My-DST on unselected MM BM aspirates with minimal perturbations ex vivo was clinically predictive of resistance to IMiDs and PIs. For the first time, this approach is able to measure the antibody-mediated cytotoxicity of daratumumab in clinical MM samples. Our data indicate that personalized regimens based on rapidly obtained ex vivo sensitivity may lead to optimized depth of clinical response. We next plan to translate this approach into clinical use in an effort to improve patient outcomes and avoid wasted time, money and complications that may arise from the use of ineffective drugs. Disclosures Mark: Janssen, Takeda, Celgene, Amgen: Consultancy; BMS, Celgene: Research Funding; Celgene: Honoraria.


2021 ◽  
Author(s):  
Yuan-Hung Wu ◽  
Yi-Ping Hung ◽  
Nai-Chi Chiu ◽  
Rheun-Chuan Lee ◽  
Chung-Pin Li ◽  
...  

Abstract BackgroundPancreatic ductal adenocarcinoma (PDAC) is highly aggressive and has poor prognosis. There are few biomarkers to inform treatment decisions, and collecting tumor samples for genomic or drug sensitivity testing is challenging.MethodsCirculating tumor cells (CTCs) were prepared from the liquid biopsies of PDAC patients. These cells were subsequently expanded ex vivo to form CTC-derived organoid cultures, using a laboratory-developed biomimetic cell culture system. The CTC-derived organoids were tested for sensitivity to a PDAC panel of nine drugs, with tests conducted in triplicate, and a weighted cytotoxicity score (CTS) was calculated from the results. Clinical response to treatment in patients was evaluated using Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 criteria at the time of blood sampling and 3 months later. CTS was then correlated with clinical response, and analyzed using 2 × 2 contingency tables.ResultsA total of 41 liquid biopsies were collected from 31 patients, with 87.8% of liquid biopsies from patients with Stage 4 disease. CTC-derived organoid expansion was achieved in 3 weeks, with 87.8% culture efficiency. CTC-derived organoid cultures were positive for EpCAM staining and negative for CD45 staining in surface marker analysis. All patients had received a median of two lines of treatment prior to enrollment, and prospective utility analysis indicated significant correlation of CTS with clinical treatment response. Two representative case studies are also presented to illustrate the relevant clinical contexts.ConclusionsIn this study, CTCs were expanded from the liquid biopsies of PDAC patients with a high success rate. Drug sensitivity profiles from CTC-derived organoid cultures correlated meaningfully with treatment response. Further studies are warranted to validate the predictive potential for this approach.Trial RegistrationTaipei Medical University Hospital Protocol Record N201803020, registered on July 10, 2018; ClinicalTrials.gov Identifier: NCT04972461, retrospectively registered on July 22, 2021.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1713-1713 ◽  
Author(s):  
Leylah Drusbosky ◽  
Taher Abbasi ◽  
Shireen Vali ◽  
Saumya Radhakrishnan ◽  
Neeraj Kumar Singh ◽  
...  

Abstract Background: Data from a small clinical trial of venetoclax in acute myeloid leukemia (AML) recently supported FDA breakthrough therapy designation for use in combination with hypomethylating agents in treatment-naïve patients who are ineligible for high-dose induction chemotherapy. Approval of this BCL-2 inhibitor raises the question of mechanism of action in AML and patient selection for treatment. Unfortunately, no biomarkers or methods exist to predict venetoclax response in AML, making treatment selection challenging. Aim: To define a novel genomic signature rule to predict AML response to venetoclax therapy and to validate the rule with ex vivo drug sensitivity testing. Methods: The Beat AML project (supported by the Leukemia & Lymphoma Society) collects clinical data and bone marrow specimens from AML patients. Bone marrow samples are analyzed by conventional cytogenetics, whole-exome sequencing, RNA-seq, and an ex vivo drug sensitivity assay. For 19 of these randomly chosen patients, every available genomic abnormality was inputted into a computational biology program (Cellworks Group) that uses PubMed and other online resources to generate patient-specific protein network maps of activated and inactivated protein pathways. Digital drug simulations with venetoclax were conducted by quantitatively measuring drug effect on a composite AML disease inhibition score (i.e., cell proliferation, viability, and apoptosis). Computational predictions of drug response were compared to venetoclax IC50 values from the Beat AML ex vivo testing. Results: Ten of the 19 AML patients were predicted by computer simulation to respond to venetoclax, and 9 of those 10 patients had the lowest IC50 values to venetoclax. Nine of the 19 patients were predicted to not respond to venetoclax, and 8 of those 9 patients had the highest IC50 values to venetoclax. Ex vivo venetoclax responses were correctly matched to their computer simulation prediction in 17 of 19 cases, and incorrectly matched in 2 cases. The positive predictive value of the computational method was 90%, negative predictive value was 89%, sensitivity was 90%, specificity was 89%, and accuracy was 89%. Amplification of the genes RB1CC1 and/or RB1was predicted by computational modeling to increase MCL1, which made venetoclax less responsive in the digital drug simulation. This genomic rule was validated with 3 AML patients: one who received venetoclax as treatment and showed refractory disease, and 2 patients who achieved complete remission after venetoclax treatment. Conclusion: We identified a new genomic signature, confirmed by functional testing, that predicts AML response to venetoclax treatment. This unique computer-based approach is intended to inform the design of phase 2 and 3 clinical trials of venetoclax in AML patients for a forthcoming precision enrollment clinical trial. Disclosures Abbasi: Cellworks: Employment. Vali:Cellworks Group: Employment. Radhakrishnan:Cellworks: Employment. Kumar Singh:Cellworks: Employment. Usmani:Cellworks: Employment. Parashar:Cellworks: Employment. Vidva:Cellworks: Employment. Druker:Agios: Honoraria; Ambit BioSciences: Consultancy; ARIAD: Patents & Royalties, Research Funding; Array: Patents & Royalties; AstraZeneca: Consultancy; Blueprint Medicines: Consultancy, Equity Ownership, Other: travel, accommodations, expenses ; BMS: Research Funding; CTI: Equity Ownership; Curis: Patents & Royalties; Cylene: Consultancy, Equity Ownership; D3 Oncology Solutions: Consultancy; Gilead Sciences: Consultancy, Other: travel, accommodations, expenses ; Lorus: Consultancy, Equity Ownership; MolecularMD: Consultancy, Equity Ownership, Patents & Royalties; Novartis: Research Funding; Oncotide Pharmaceuticals: Research Funding; Pfizer: Patents & Royalties; Roche: Consultancy.


Hemato ◽  
2021 ◽  
Vol 2 (2) ◽  
pp. 167-181
Author(s):  
Marie Thérèse Rubio ◽  
Adèle Dhuyser ◽  
Stéphanie Nguyen

Myeloma tumor cells are particularly dependent on their microenvironment and sensitive to cellular antitumor immune response, including natural killer (NK) cells. These later are essential innate lymphocytes implicated in the control of viral infections and cancers. Their cytotoxic activity is regulated by a balance between activating and inhibitory signals resulting from the complex interaction of surface receptors and their respective ligands. Myeloma disease evolution is associated with a progressive alteration of NK cell number, phenotype and cytotoxic functions. We review here the different therapeutic approaches that could restore or enhance NK cell functions in multiple myeloma. First, conventional treatments (immunomodulatory drugs-IMids and proteasome inhibitors) can enhance NK killing of tumor cells by modulating the expression of NK receptors and their corresponding ligands on NK and myeloma cells, respectively. Because of their ability to kill by antibody-dependent cell cytotoxicity, NK cells are important effectors involved in the efficacy of anti-myeloma monoclonal antibodies targeting the tumor antigens CD38, CS1 or BCMA. These complementary mechanisms support the more recent therapeutic combination of IMids or proteasome inhibitors to monoclonal antibodies. We finally discuss the ongoing development of new NK cell-based immunotherapies, such as ex vivo expanded killer cell immunoglobulin-like receptors (KIR)-mismatched NK cells, chimeric antigen receptors (CAR)-NK cells, check point and KIR inhibitors.


2014 ◽  
Vol 2014 ◽  
pp. 1-8 ◽  
Author(s):  
Youngil Koh ◽  
Woo-June Jung ◽  
Kwang-Sung Ahn ◽  
Sung-Soo Yoon

Purpose.We tried to establish clinically relevant human myeloma cell lines that can contribute to the understanding of multiple myeloma (MM).Materials and Methods.Mononuclear cells obtained from MM patient’s bone marrow were injected via tail vein in an NRG/SCID mouse. Fourteen weeks after the injection, tumor developed at subcutis of the mouse. The engraftment of MM cells into mouse bone marrow (BM) was also observed. We separated and cultured cells from subcutis and BM.Results.After the separation and culture of cells from subcutis and BM, we established two cell lines originating from a single patient (SNU_MM1393_BM and SNU_MM1393_SC). Karyotype of the two newly established MM cell lines showed tetraploidy which is different from the karyotype of the patient (diploidy) indicating clonal evolution. In contrast to SNU_MM1393_BM, cell proliferation of SNU_MM1393_SC was IL-6 independent. SNU_MM1393_BM and SNU_MM1393_SC showed high degree of resistance against bortezomib compared to U266 cell line. SNU_MM1393_BM had the greater lethality compared to SNU_MM1393_SC.Conclusion.Two cell lines harboring different site tropisms established from a single patient showed differences in cytokine response and lethality. Our newly established cell lines could be used as a tool to understand the biology of multiple myeloma.


Sign in / Sign up

Export Citation Format

Share Document