scholarly journals A Rapid Functional Screen for Small Molecule and Monoclonal Antibody Drug Sensitivity in Multiple Myeloma Patients

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 ◽  
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.


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 &lt;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.


2015 ◽  
Vol 15 ◽  
pp. e224-e225 ◽  
Author(s):  
J.M. Waldschmidt ◽  
D. Wider ◽  
A.R. Thomsen ◽  
C. Aldrian ◽  
A. Simon ◽  
...  

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.


Author(s):  
Cristina E. Tognon ◽  
Rosalie C. Sears ◽  
Gordon B. Mills ◽  
Joe W. Gray ◽  
Jeffrey W. Tyner

The use of ex vivo drug sensitivity testing to predict drug activity in individual patients has been actively explored for almost 50 years without delivering a generally useful predictive capability. However, extended failure should not be an indicator of futility. This is especially true in cancer research, where ultimate success is often preceded by less successful attempts. For example, both immune- and genetic-based targeted therapies for cancer underwent numerous failed attempts before biological understanding, improved targets, and optimized drug development matured to facilitate an arsenal of transformational drugs. Similarly, directly assessing drug sensitivity of primary tumor biopsies—and the use of this information to help direct therapeutic approaches—has a long history with a definitive learning curve. In this review, we survey the history of ex vivo testing and the current state of the art for this field. We present an update on methodologies and approaches, describe the use of these technologies to test cutting-edge drug classes, and describe an increasingly nuanced understanding of tumor types and models for which this strategy is most likely to succeed. We consider the relative strengths and weaknesses of predicting drug activity across the broad biological context of cancer patients and tumor types. This includes an analysis of the potential for ex vivo drug sensitivity testing to accurately predict drug activity within each of the biological hallmarks of cancer pathogenesis. Expected final online publication date for the Annual Review of Cancer Biology, Volume 5 is March 4, 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 37-38
Author(s):  
Olivia Perez De Acha ◽  
Beau M Idler ◽  
Zachary J Walker ◽  
Peter A Forsberg ◽  
Tomer M Mark ◽  
...  

Background: Daratumumab-refractory multiple myeloma (MM) patients have limited treatment options and a dire prognosis. Daratumumab (Dara) targets the overexpressed myeloma antigen CD38, and its mechanism of resistance has been partially associated with downregulation of CD38 expression (Nijhof et al. Blood, 2016). Dara is a key agent in the relapsed setting and is being integrated into upfront treatment. In addition, isatuximab (Isa) has become the second FDA-approved anti-CD38 monoclonal antibody (mAb). Thus, when and how patients can be retreated with this important drug-class has become a critical question. We developed a platform for measuring drug efficacy ex vivo, including mAbs, termed Myeloma Drug Sensitivity Testing (My-DST) (Walker et al. Blood Advances, 2020). Here, we addressed whether My-DST could determine if retreatment with the anti-CD38 mAbs Dara and Isa can be effective in patients that had previously developed Dara-resistance. Methods: Bone marrow aspirates were obtained from the hematologic tissue bank (HTB) from patients at the University of Colorado Blood Cancer Program after informed consent and IRB approval. Mononuclear cells (MNCs) were isolated by Ficoll density gradient centrifugation, cells incubated for 48 h with 20 nM Dara, Isa and untreated controls in triplicate wells. Flow cytometry was performed on a BD FACSCelesta. To identify the viable MM population, samples were stained with fluorophore-conjugated mAbs to CD38, CD138, CD45, CD19, CD56 and CD46. Stained samples were washed and resuspended in Live/Dead dye Near-IR. Results were analyzed through FlowJo and Graphpad Prism software. Results: My-DST for Dara (20 nM) cytotoxicity has been performed in 67 patients, 52 of which were Dara-naïve and 15 of which were clinically Dara-refractory. Of Dara-naïve patients, 38/52 (73%) showed &gt;20% reduction in viable MM cells whereas only 3/15 (20%) of the Dara-refractory patients showed &gt;20% decrease (Fig 1A). Provocatively, the patients that showed potential "re-sensitization" had been off Dara for &gt;12 months, and there was a significant correlation between response to Dara and months off treatment (r= -0.5096, p= 0.0457, Fig 1B). Furthermore, we evaluated two timepoints in one of the responders (HTB1749), and the later sample (HTB1749.3) showed a deeper response, further supporting the correlation. Surprisingly, in Dara-refractory patients the median CD38 expression was not significantly different between the three ex vivo responders and those who did not respond (p = 0.439, Fig 1C). Isa was tested in seven of the Dara-refractory patient samples, showing &gt;20% decrease in viable MM cells in five, of which three did not respond ex vivo to Dara (Fig 1D). Interestingly, the Isa responders were only off Dara treatment for an average of 7.5 months. We further investigated HTB1059.3 and HTB1749.2, which responded to Dara, and found that these patients had multiple distinct MM cell subpopulations with different levels of CD38 expression, with the subpopulation with higher CD38 MFI accounting for the decreased viability (Fig 1E-F). When we compared HTB1059.3 with a prior sample available, these differential populations were not present before Dara treatment (data not shown), indicating these subpopulations evolved after treatment. Likewise, the same phenomenon of subpopulations with differential CD38 expression was observed in the Dara-refractory patients who responded to Isa (Fig 1G-H). Conclusion: These data support the possibility of retreatment with anti-CD38 mAbs in patients who once became refractory to Dara. Although our findings need to be confirmed with additional samples, they suggest that Isa may have efficacy earlier in this setting, supporting an approach to switch agents when retreating with this drug class. Anti-CD38 mAb sensitivity in the Dara-refractory population appears to be heavily influenced by the different CD38 expression levels on the heterogeneous MM cell subpopulations that emerge when a patient is off Dara for a period of time. Furthermore, My-DST with anti-CD38 mAbs may be applied to help guide the treatment approach in this population. Still, the presence of CD38-low subpopulations in these patients may represent resistant cells that shorten the remission times on retreatment. Thus, the combination drug partner choice will likely be critical to successfully optimizing depth and duration of response in Dara-refractory patients. Figure 1 Disclosures Forsberg: Genentech, Inc., Sanofi, Karyopharm, Abbvie: Research Funding; Celgene: Speakers Bureau. Mark:Bristol-Myers Squibb: Research Funding; Janssen: Research Funding; Celgene: Consultancy; Amgen: Consultancy; Kayopharm: Consultancy; Janssen: Consultancy; Sanofi: Consultancy; Takeda: Consultancy. Sherbenou:Oncopeptides Inc.: Research Funding.


2019 ◽  
Author(s):  
Sergey G. Kuznetsov ◽  
Alexander Ianevski ◽  
Evgeny Kulessky ◽  
Karoliina Laamanen ◽  
Elina Lehtinen ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4770-4770
Author(s):  
Anna C. Schinzel ◽  
Mike Chapman ◽  
Ben Wittner ◽  
Yashaswi Shrestha ◽  
Sridhar Ramaswamy ◽  
...  

Abstract Karyotypic complexity is one of the hallmarks of multiple myeloma, complicating efforts to identify the specific genetic alterations involved in the development of multiple myeloma. To identify genes essential for myeloma proliferation, we used a high-throughput loss-of-function approach using lentivirally delivered short hairpin RNA (shRNA) library targeting human kinases as part of a Multiple Myeloma Research Foundation sponsored pilot project. Three myeloma cell lines, MM.1S, RPMI8226 and INA6, were infected in an arrayed 384-well format. All infections were performed in quadruplicate; two replicates were treated with puromycin to select for infected cells and two were left untreated. Wells in which more than 25% of the cells survived puromycin treatment, as determined by a cell viability assay, were included in future analysis. Genes in which 2 or more hairpins scored below a defined threshold were considered as ‘hits’. To identify physiologically relevant hits, we have integrated these findings with whole genome analysis of copy number and gene expression of multiple myeloma. We have identified a number of genes that are both amplified in and required for multiple myeloma cell viability. These observations suggest potential new targets for therapeutic targeting in myeloma.


Sign in / Sign up

Export Citation Format

Share Document