Autophagic degradation of NOXA underlies stromal cell-mediated resistance to proteasome inhibitors in mantle cell lymphoma

2021 ◽  
pp. 106672
Author(s):  
Yoshiaki Kuroda ◽  
Daisuke Koyama ◽  
Jiro Kikuchi ◽  
Shigehisa Mori ◽  
Tatsuo Ichinohe ◽  
...  
Leukemia ◽  
2020 ◽  
Author(s):  
Yoshiaki Kuroda ◽  
Akiko Yashima-Abo ◽  
Daisuke Koyama ◽  
Jiro Kikuchi ◽  
Shigehisa Mori ◽  
...  

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 17522-17522 ◽  
Author(s):  
J. Drach ◽  
H. Kaufmann ◽  
O. Pichelmayer ◽  
V. Sagaster ◽  
S. Seidl ◽  
...  

17522 Background: Bortezomib (B) belongs to a new class of anti-cancer agents, the proteasome inhibitors, and has documented activity in multiple myeloma and mantle cell lymphoma (MCL). Preclinical studies suggest that B has synergistic activity with rituximab (R), which provides a rationale for the exploration of treatment combinations. Methods: We have initiated a phase II study in relapsed/chemotherapy refractory MCL to evaluate the activity and safety of B in combination with R and dexamethasone (BORID). A treatment cycle consists of B at 1.3 mg/m2 administered on days 1, 4, 8, and 11, R at 375 mg/m2 administered on day 1, and dexamethasone 40 mg orally on days 1 to 4. Cycles are repeated every 3 weeks for a total of 6 treatment cycles. Patients (pts) with progressive MCL after at least one prior line of therapy (including CHOP or a CHOP-like regimen) are eligible. Results: Up to now, we have enrolled 10 pts (median age, 69 years; range, 48 to 75 years) after a median of 3 lines of prior therapies (range, 1 to 6) including R in 8 pts, high-dose therapy in 3 pts, and thalidomide in 5 pts. Median time between start of frontline therapy and study inclusion was 43 months (range, 11 to 98 months). Severe adverse events (> grade II) included infections (herpes zoster in 2 pts, bacterial pneumonia, mucosal candidiasis), peripheral neuropathy (3 pts), fatigue (2 pts) and vasculitic skin infiltrates in 3 pts. Thrombopenia (< 50 G/L) occured in 2 pts. All adverse events were managable by standard means of supportive care and prolongation of the treatment interval between cycles. Of 8 pts evaluable for efficacy, 7 have achieved a response (3 CR, 4 PR), and 1 pt experienced stable disease. Pts in CR were also negative for disease activity by PET scanning. Skin infiltrates (histologically proven T-cell infiltrates) preceded achievement of CR in 2 pts. 6 of 6 pts are still progression-free at 6 months after treatment initiation. Recruitment of patients is ongoing, and updated results will be presented. Conclusions: Data obtained thus far indicate that BORID has promising activitiy and managable toxicity in patients with heavily pretreated MCL, and development of a vasculitic rash may be an early indicator of a favorable response. [Table: see text]


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2532-2532 ◽  
Author(s):  
Victor Y. Yazbeck ◽  
Georgios V. Georgakis ◽  
Yang Li ◽  
David McConkey ◽  
Michael Andreeff ◽  
...  

Abstract Mantle cell lymphoma (MCL) accounts for 6–8% of all non-Hodgkin lymphomas (NHLs). It is an aggressive lymphoma with a poor prognosis--it is generally considered incurable with conventional treatments, and median survival is 3–4 years with a 10-year survival of only 10–15%. There is no accepted standard of care and effective treatments are greatly needed. Bcl-2 family proteins are important regulators of the intrinsic apoptotic pathway and are involved in oncogenesis and chemoresistance of a variety of tumor types, including lymphoma. Antiapoptotic proteins of the Bcl-2 family are overexpressed in mantle cell lymphoma (MCL) cells and may be responsible, in part, for drug resistance. GX15-070 is a small-molecule antagonist of the BH3-binding groove of the Bcl-2 family of proteins, and is currently in Phase I clinical trials. Consequently, we determined the activity of GX15-070 in 3 MCL cell lines (Jeko-1, Mino, and SP53). Cell viability was determined by MTS assay, apoptosis by Annexin-V binding and FACS analysis, and molecular changes by western blot. GX15-070 induced apoptosis in all three MCL cell lines in a dose and time-dependent manner. In the SP53 cell line, GX15-070 decreased MCL-1 and Bak levels, increased Bax and cleaved caspase 3. Furthermore, GX15-070 activated both the extrinsic and intrinsic apoptotic pathway as evident by cleavage of caspase 8, 9, and Bid. Both bortezomib and the novel proteasome inhibitor NPI-0052 induced single agent antiproliferative activity in MCL. GX15-070 enhanced the effect of both proteasome inhibitors. Additionally, GX15-070 showed an additive effect with doxorubicin. These studies suggest that GX15-070 may have a therapeutic value in MCL either alone or in combination with proteasome inhibitors or chemotherapy.


Hematology ◽  
2007 ◽  
Vol 2007 (1) ◽  
pp. 270-276 ◽  
Author(s):  
Owen A. O’Connor

Abstract Mantle cell lymphoma (MCL) remains one of the more challenging sub-types of non-Hodgkin lymphoma. This entity, which is only approximately 10 years old, is characterized by response to many different chemotherapy regimens, though the duration of those responses remains often times quite short. Retreatment with second and third line combination regimens results in shorter and shorter durations of response, with the rapid emergence of a very drug-resistant phenotype. Despite these often frustrating clinical features, there is now a lot of new hope in managing patients with MCL. New insights into the molecular pathogenesis of MCL has revealed a plethora of new potential targets, while our continued efforts in novel targeted drug development has produced a host of agents that are already helping patients with this challenging disease. The use of proteasome inhibitors, for example, represents one example of a new strategy that has offered new hope for patients, and new opportunities for the physician treating this disease. In this review, we will put this biology into perspective, and describe how new revelations in MCL pathogenesis are leading to the identification of many exciting new drugs with promising activity.


Blood ◽  
2010 ◽  
Vol 116 (20) ◽  
pp. 4185-4191 ◽  
Author(s):  
Sarah K. Brennan ◽  
Brooke Meade ◽  
Qiuju Wang ◽  
Akil A. Merchant ◽  
Jeanne Kowalski ◽  
...  

AbstractPatients with mantle cell lymphoma (MCL) typically respond to initial treatment but subsequently relapse. This pattern suggests that a population of MCL cells is both drug resistant and capable of clonogenic growth. The intracellular enzyme retinaldehyde dehydrogenase (ALDH) provides resistance to several toxic agents. ALDH can also identify stem cells in normal adult tissues and tumorigenic cancer stem cells in several human malignancies. We studied ALDH expression in MCL and found small populations of ALDH+ cells that were highly clonogenic. Moreover, ALDH+ MCL cells were relatively quiescent and resistant to a wide range of agents. Normal B cells can be activated by specific unmethylated cytosine-phosphate-guanosine (CpG) DNA motifs through toll-like receptor 9, and we found that the synthetic CpG oligonucleotide 2006 (CpG) reduced the frequency of quiescent ALDH+ MCL cells, induced terminal plasma cell differentiation, and limited tumor formation in vitro and in vivo. Treatment with CpG also significantly enhanced the activity of the proteasome inhibitor bortezomib that was associated with induction of the unfolded protein response. Our data suggest that CpG may target clonogenic and resistant ALDH+ cells as well as improve the activity of proteasome inhibitors in MCL.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1490-1490
Author(s):  
Patricia Perez Galan ◽  
Gael Roue ◽  
Neus Villamor ◽  
Elias Campo ◽  
Dolors Colomer

Abstract Mantle cell lymphoma (MCL) is a lymphoid malignancy derived from a subset of mature B-cells with coexpression of CD5 and the chromosomal translocation t(11;14)(q13;q32). MCL patients present an aggressive clinical course and poor response to conventional chemotherapy due to either rapid relapse after an initial response or primary resistance to drugs. Thus, there is currently a strong effort to develop compounds that target novel biological pathways. In addition, MCL cells overexpress the antiapoptotic proteins Bcl-2, Bcl-XL and Mcl-1. The dysregulated Bcl-2 pathway represents an important target for a new-mechanism therapeutic approach. In this context, the pan-Bcl-2 inhibitor GX15-070 that belongs to a new family of compounds which mimic BH3-only proteins by binding to multiple antiapoptotic Bcl-2 members and consequently eliciting Bax and Bak dependent cytotoxicity. GX15-070 is currently being evaluated in Phase I clinical trials. GX15-070 induced apoptosis in vitro in primary cells from MCL patients as well in MCL cells lines, at doses ranging from 0,5μM to 5μM. GX15-070 induced phosphatidylserine exposure, mitochondrial depolarization, ROS generation, Bax and Bak conformational changes and caspase activation. As expected, pan-caspase inhibitors did not block apoptosis in accordance with the mitochondria-targeted nature of this new compound. The susceptibility of MCL cells correlates with the expression of Bcl-2, Mcl-1 and Bcl-XL, both at protein and mRNA levels. Although GX15-070 exhibited significant in vitro cytotoxicity in MCL as a single agent, we tested also this compound in combination with proteasome inhibitors. Bortezomib (PS-341, Velcade®) has shown promising results in MCL treatment. One of the drawbacks of proteasome inhibitors is the undesirable accumulation, due to absence of degradation, of antiapoptotic factors which could impede or slow apoptosis, such as Mcl-1 protein. With this rationale we evaluated a combination of bortezomib with the pan-Bcl-2 inhibitor GX15-070 in vitro in MCL cells. Preincubation of MCL cells with GX15-070 at doses ranging from 0,1μM to 1μM before bortezomib (1–10nM) addition induced a synergistic cytotoxic effect (Chou Talalay CI =[0,26–0,575]). In vitro, this new combination allows to reduce bortezomib dose 5 to 20 times, depending on the individuals. Western blot analysis revealed that this combination reduced Mcl-1 levels and displaced Bak from Mcl-1 sequestration. Moreover, upregulation of the proapoptotic BH3-only protein Noxa by bortezomib, was further increased when combined with GX15-070. All these findings suggest that GX15-070 and bortezomib combination could be a new therapeutic approach for MCL treatment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2825-2825 ◽  
Author(s):  
Antonina Kurtova ◽  
Archie Tamayo ◽  
Richard J. Ford ◽  
Jan A. Burger

Abstract Mantle cell lymphoma (MCL) is an aggressive lymphoma that generally is associated with a rapid dissemination of the malignant B-cells and a high risk of relapse. Chemokine receptors and adhesion molecules play key roles in normal B cell migration and homing to distinct microenvironments, but their expression and function in MCL is largely unknown. In this study, we profiled the expression and function of chemokine receptors and adhesion molecules (CXCR4, CXCR5, CXCR3, CD49d/VLA-4, CD44, and CD62L) in MCL cell lines (SP-53, MINO, JeKo-1 and Granta-519) and primary MCL cells. Except for the EBV-positive cell line Granta 519, all MCL lines displayed high levels of CXCR4, CXCR5, CD49d and CD44. Primary MCL cells from different patients (n=6) displayed a similar immunophenotype. We then analyzed chemotaxis of MCL cells towards CXCL12 (200 ng/ml) and CXCL13 (1μg/ml) in transwell assays. 35.7±5.7% of input SP-53, 25.8±2.8% of MINO, and 6.7±0.9% of JeKo-1 cells migrated towards CXCL12 within 3 hours (mean±SEM, triplicates). 43.4±9.8% of input SP-53, 16.7±2.8% of MINO, and 4.3±0.5% of JeKo-1 cells migrated toward CXCL13. Granta-519 did not demonstrate any significant chemotaxis. Pre-treating the cells with a CXCR4 antagonist (AMD3100, Plerixafor) effectively blocked chemotaxis towards CXCL12. Marrow stromal cells (MSC) constitutively secrete CXCL12 and provide ligands for VLA-4 integrins. These two molecules now can be targeted clinically using CXCR4 antagonists or monoclonal antibodies (mAbs), respectively. In co-cultures with MSC, SP-53 and MINO displayed abundant spontaneous migration beneath MSC (pseudoemperipolesis/PEP). Therefore we examined whether blocking of CXCR4 or VLA-4 affects the PEP of MCL cells. Pre-incubation of SP-53 or MINO cells with AMD3100 reduced PEP to levels that were 49.4±1.5% (p&lt;.07) or 11.7±1.7% (p&lt;.04) of controls. Pretreatment with anti-VLA-4 antibodies (Natalizumab) also resulted in significant decrease of PEP to levels that were 5.8±1.4% (p&lt;.02) in SP-53 and 2.2±0.6% (p&lt;.01) in MINO cells. Pre-incubation with a cyclic peptide inhibitor with the minimal VLA-4 binding motif “LDV” also significantly reduced PEP of MCL cells to 22.1±4.0% (p&lt;.006) in SP-53 and 7.1±1.9% (p&lt;.005) in MINO cells. MCL cells treated with 10 μM fludarabine (F-ara-A) in suspension cultures resulted in high levels of apoptosis in MCL cells within 24 to 72 hrs. We found that co-culture with MSC significantly reduced F-ara-A-induced apoptosis, a primary drug resistance mechanism termed cell adhesion-mediated drug resistance (CAM-DR). Interestingly, the MCL cell fraction that had migrated into the stromal cell layer was particularly protected from the cytotoxic effect of F-ara-A. As such, clinical targeting of CXCR4 and VLA-4 integrins in MCL may not only antagonize the migration and homing associated with the dissemination of the disease, but also disrupt the adhesion to stromal cells, help to overcome CAM-DR, and thus make MCL cells more accessible to conventional drugs. Collectively, our studies provide a rationale to further explore the efficacy of combinations of CXCR4- and/or VLA-4 antagonists with conventional drugs in patients with this disease. This approach may lead to new therapeutic avenues for MCL patients.


Cancers ◽  
2020 ◽  
Vol 12 (6) ◽  
pp. 1565
Author(s):  
Gaël Roué ◽  
Brigitte Sola

Mantle cell lymphoma (MCL) is a rare but aggressive B-cell hemopathy characterized by the translocation t(11;14)(q13;q32) that leads to the overexpression of the cell cycle regulatory protein cyclin D1. This translocation is the initial event of the lymphomagenesis, but tumor cells can acquire additional alterations allowing the progression of the disease with a more aggressive phenotype and a tight dependency on microenvironment signaling. To date, the chemotherapeutic-based standard care is largely inefficient and despite the recent advent of different targeted therapies including proteasome inhibitors, immunomodulatory drugs, tyrosine kinase inhibitors, relapses are frequent and are generally related to a dismal prognosis. As a result, MCL remains an incurable disease. In this review, we will present the molecular mechanisms of drug resistance learned from both preclinical and clinical experiences in MCL, detailing the main tumor intrinsic processes and signaling pathways associated to therapeutic drug escape. We will also discuss the possibility to counteract the acquisition of drug refractoriness through the design of more efficient strategies, with an emphasis on the most recent combination approaches.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1541-1541
Author(s):  
Jonathon B. Cohen ◽  
Craig A. Portell ◽  
Mehdi Hamadani ◽  
Opeyemi Jegede ◽  
Catherine Diefenbach ◽  
...  

Background: The Bruton's tyrosine kinase inhibitor ibrutinib is highly effective as a monotherapy in relapsed/refractory mantle cell lymphoma (MCL) with an overall response rate of 68% (Wang et al, NEJM 2013), but the duration of response is shorter than what is seen in chronic lymphocytic leukemia, and the survival of patients who progress after receiving ibrutinib is as short as 3 months (Martin et al, Blood, 2016). In addition, the complete response (CR) rate is only 21%. Ibrutinib-containing combinations may improve depth and duration of response in patients with relapsed/refractory MCL. While use of the proteasome inhibitor, bortezomib, can be limited due to the development of peripheral neuropathy, it has an ORR of 33% (CR rate 8%) in MCL, and preclinical models suggest a synergism between proteasome inhibitors and ibrutinib in MCL cell lines (Axelrod et al, Leukemia 2014). We developed a phase 1/2 trial of ibrutinib combined with the oral proteasome inhibitor ixazomib in patients with relapsed/refractory MCL. Methods: PrE0404 will be open at 18 sites nationwide and is registered at clinicaltrials.gov (NCT03323151). It is currently enrolling patients with relapsed/refractory MCL who have received at least 1 prior line of combination therapy. Patients receiving prior BTK or proteasome inhibitors are eligible, and patients may have received prior autologous or allogeneic transplantation as long as they do not have active graft versus host disease. Patients must have ≤ grade 1 peripheral neuropathy. For phase 1, patients are required to have been off of a BTK inhibitor for 3 months. Starting dose of ibrutinib for all patients is 560mg daily, and dose levels of ixazomib for the phase 1 trial range from 3mg to 4mg days 1, 8, and 15 of a 28 day cycle. Patients continued therapy until disease progression or unacceptable toxicity. For the phase 1 portion of the study, patients are monitored for a dose limiting toxicity (DLT) during cycle 1, defined as grade 3 thrombocytopenia with significant bleeding, select grade 3 non-hematologic toxicities, grade 4 thrombocytopenia, grade 4 febrile neutropenia, grade 4 non-hematologic toxicity, or any grade 5 toxicity. In addition, any toxicity-related dose delay &gt; 7 days of ibrutinib or ixazomib or an inability to receive all 3 doses of ixazomib during cycle 1 are considered DLT's. The maximum tolerated dose/recommended phase 2 dose will be the dose at which fewer than 1/6 patients experience a DLT, with the maximum dose of ixazomib will be 4mg. The primary endpoint for the phase 2 portion of the study is CR rate, and patients will be assigned to one of two cohorts based on prior BTK-inhibitor exposure. For ibrutinib-naïve patients, we will target a CR rate of 40% (based on a historical CR rate of 21% for ibrutinib), and for ibrutinib-pretreated patients, we will target a CR rate of 23% (based on a historical CR rate of 8% for bortezomib). There is 86% statistical power & a one-sided 10% alpha to test each hypothesis. We will accrue 31 patients to each cohort in order to detect this difference. Secondary and exploratory endpoints will include progression-free and overall survival, overall response, toxicity, frequency of BTK mutations, and response based on molecular risk stratification. As of July 2019 the study is open to accrual at 14 sites and is expected to move to phase 2 in fall 2019, at which time it will be expanded to 18 sites. Disclosures Cohen: Hutchison: Research Funding; Seattle Genetics, Inc.: Consultancy, Research Funding; Bristol-Meyers Squibb Company: Research Funding; Genentech, Inc.: Consultancy, Research Funding; Janssen Pharmaceuticals: Consultancy; Astra Zeneca: Research Funding; LAM Therapeutics: Research Funding; Lymphoma Research Foundation: Research Funding; ASH: Research Funding; UNUM: Research Funding; Gilead/Kite: Consultancy; Takeda Pharmaceuticals North America, Inc.: Research Funding. Portell:Infinity: Research Funding; Roche/Genentech: Research Funding; Xencor: Research Funding; TG Therapeutics: Research Funding; Acerta/AstraZeneca: Research Funding; Kite: Consultancy, Research Funding; Bayer: Consultancy; AbbVie: Research Funding; Pharmacyclics: Consultancy; Janssen: Consultancy; Genentech: Consultancy, Research Funding; Amgen: Consultancy; BeiGene: Consultancy, Research Funding. Hamadani:ADC Therapeutics: Consultancy, Research Funding; Janssen: Consultancy; Sanofi Genzyme: Research Funding, Speakers Bureau; Otsuka: Research Funding; Celgene: Consultancy; Merck: Research Funding; Medimmune: Consultancy, Research Funding; Takeda: Research Funding; Pharmacyclics: Consultancy. Diefenbach:Bristol-Myers Squibb: Consultancy, Research Funding; Denovo: Research Funding; Genentech: Consultancy, Research Funding; Incyte: Research Funding; LAM Therapeutics: Research Funding; MEI: Research Funding; Merck: Consultancy, Research Funding; Seattle Genetics: Consultancy, Research Funding; Millenium/Takeda: Research Funding; Trillium: Research Funding. Landsburg:Celgene: Membership on an entity's Board of Directors or advisory committees; Triphase: Research Funding; Triphase: Research Funding; Takeda: Research Funding; Takeda: Research Funding; Seattle Genetics: Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees; Curis, INC: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Curis, INC: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Seattle Genetics: Speakers Bureau. Kahl:Seattle Genetics: Consultancy; ADC Therapeutics: Consultancy, Research Funding; BeiGene: Consultancy; TG Therapeutics: Consultancy. OffLabel Disclosure: Ixazomib is not currently approved for mantle cell lymphoma.


2018 ◽  
Vol 17 (5) ◽  
pp. 1090-1100 ◽  
Author(s):  
Jiyu Guan ◽  
Dan Huang ◽  
Konstantin Yakimchuk ◽  
Sam Okret

Sign in / Sign up

Export Citation Format

Share Document