17 results on '"Daniel D Waller"'
Search Results
2. Data from IL6 Promotes a STAT3-PRL3 Feedforward Loop via SHP2 Repression in Multiple Myeloma
- Author
-
Wee-Joo Chng, Michael Sebag, Daniel D. Waller, Qi Zeng, Zea Tuan Tan, Tae-Hoon Chung, Jing-Yuan Chooi, Yan Ting Hee, Julia S.L. Lim, Jianbiao Zhou, and Phyllis S.Y. Chong
- Abstract
Overexpression of PRL-3, an oncogenic phosphatase, was identified as a novel cluster in patients with newly diagnosed multiple myeloma. However, the regulation and oncogenic activities of PRL-3 in multiple myeloma warrant further investigation. Here, we report that IL6 activates STAT3, which acts as a direct transcriptional regulator of PRL-3. Upregulation of PRL-3 increased myeloma cell viability and rephosphorylated STAT3 in a biphasic manner through direct interaction and deactivation of SHP2, thus blocking the gp130 (Y759)-mediated repression of STAT3 activity. Abrogation of PRL-3 reduced myeloma cell survival, clonogenicity, and tumorigenesis, and detailed mechanistic studies revealed “deactivation” of effector proteins such as Akt, Erk1/2, Src, STAT1, and STAT3. Furthermore, loss of PRL-3 efficiently abolished nuclear localization of STAT3 and reduced its occupancy on the promoter of target genes c-Myc and Mcl-1, and antiapoptotic genes Bcl2 and Bcl-xL. PRL-3 also played a role in the acquired resistance of myeloma cells to bortezomib, which could be overcome by PRL-3 silencing. Of clinical relevance, STAT3 and PRL-3 expression was positively correlated in five independent cohorts, and the STAT3 activation signature was significantly enriched in patients with high PRL-3 expression. Furthermore, PRL-3 could be used as a biomarker to identify high-risk patients with multiple myeloma that exhibited poor prognosis and inferior outcome even when treated with novel combinational therapeutics (proteasome inhibitors and immunomodulatory imide drugs). Conclusively, our results support a feedforward mechanism between STAT3 and PRL-3 that prolongs prosurvival signaling in multiple myeloma, and suggest targeting PRL-3 as a valid therapeutic opportunity in multiple myeloma.Significance:IL6 promotes STAT3-dependent transcriptional upregulation of PRL-3, which in turn re-phosphorylates STAT3 and aberrantly activates STAT3 target genes, leading to bortezomib resistance in multiple myeloma.
- Published
- 2023
3. Synthesis and Evaluation of Structurally Diverse C-2-Substituted Thienopyrimidine-Based Inhibitors of the Human Geranylgeranyl Pyrophosphate Synthase
- Author
-
Hiu-Fung Lee, Cyrus M. Lacbay, Rebecca Boutin, Alexios N. Matralis, Jaeok Park, Daniel D. Waller, Tian Lai Guan, Michael Sebag, and Youla S. Tsantrizos
- Subjects
Male ,Molecular Structure ,Antineoplastic Agents ,Bone Marrow Cells ,Saccharomyces cerevisiae ,Thiophenes ,Rats ,Fungal Proteins ,Mice, Inbred C57BL ,Structure-Activity Relationship ,Pyrimidines ,Liver ,Geranylgeranyl-Diphosphate Geranylgeranyltransferase ,Drug Discovery ,Animals ,Humans ,Molecular Medicine ,Female ,Enzyme Inhibitors ,Multiple Myeloma ,Protein Binding - Abstract
Novel analogues of C-2-substituted thienopyrimidine-based bisphosphonates (C2-ThP-BPs) are described that are potent inhibitors of the human geranylgeranyl pyrophosphate synthase (hGGPPS). Members of this class of compounds induce target-selective apoptosis of multiple myeloma (MM) cells and exhibit antimyeloma activity
- Published
- 2022
4. Longitudinal single-cell analysis of a myeloma mouse model identifies subclonal molecular programs associated with progression
- Author
-
Marta Chesi, Laura M. Richards, Daniel D Waller, Serges P Tsofack, Ellen Nong Wei, P. Leif Bergsagel, Suzanne Trudel, Danielle C Croucher, Xian Fang Huang, Michael Sebag, Trevor J. Pugh, and Zhihua Li
- Subjects
DNA Copy Number Variations ,Tumour heterogeneity ,Science ,General Physics and Astronomy ,Myeloma ,Disease ,Protein Serine-Threonine Kinases ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Genetic Heterogeneity ,Mice ,Targeted therapies ,Single-cell analysis ,Cancer genomics ,medicine ,Animals ,Humans ,Copy-number variation ,Multiple myeloma ,Multidisciplinary ,Mechanism (biology) ,Disease progression ,General Chemistry ,medicine.disease ,In vitro ,Mice, Inbred C57BL ,Disease Models, Animal ,Disease Progression ,Cancer research ,Single-Cell Analysis ,Multiple Myeloma - Abstract
Molecular programs that underlie precursor progression in multiple myeloma are incompletely understood. Here, we report a disease spectrum-spanning, single-cell analysis of the Vκ*MYC myeloma mouse model. Using samples obtained from mice with serologically undetectable disease, we identify malignant cells as early as 30 weeks of age and show that these tumours contain subclonal copy number variations that persist throughout progression. We detect intratumoural heterogeneity driven by transcriptional variability during active disease and show that subclonal expression programs are enriched at different times throughout early disease. We then show how one subclonal program related to GCN2 stress response is progressively activated during progression in myeloma patients. Finally, we use chemical and genetic perturbation of GCN2 in vitro to support this pathway as a therapeutic target in myeloma. These findings therefore present a model of precursor progression in Vκ*MYC mice, nominate an adaptive mechanism important for myeloma survival, and highlight the need for single-cell analyses to understand the biological underpinnings of disease progression., The molecular programs that underlie progression in multiple myeloma (MM) are incompletely understood. Here the authors use a mouse model of MM and single-cell RNA-seq to define subclonal expression programs that arise during progression and that inform targeted therapeutic strategies.
- Published
- 2021
5. CLIC-01: Manufacture and distribution of non-cryopreserved CAR-T cells for patients with CD19 positive hematologic malignancies
- Author
-
Natasha Kekre, Kevin A. Hay, John R. Webb, Ranjeeta Mallick, Miruna Balasundaram, Mhairi K. Sigrist, Anne-Marie Clement, Julie S. Nielsen, Jennifer Quizi, Eric Yung, Scott D. Brown, Lisa Dreolini, Daniel D. Waller, Julian Smazynski, Nicole S. Gierc, Bianca C. Loveless, Kayla Clark, Tyler Dyer, Richard Hogg, Leah McCormick, Michael Gignac, Shanti Bell, D. Maria Chapman, David Bond, Siao Yong, Rachel Fung, Heather M. Lockyer, Victoria Hodgson, Catherine Murphy, Ana Subramanian, Evelyn Wiebe, Piriya Yoganathan, Liana Medynski, Dominique C. Vaillan, Alice Black, Sheryl McDiarmid, Michael Kennah, Linda Hamelin, Kevin Song, Sujaatha Narayanan, Judith A. Rodrigo, Stefany Dupont, Terry Hawrysh, Justin Presseau, Kednapa Thavorn, Manoj M. Lalu, Dean A. Fergusson, John C. Bell, Harold Atkins, Brad H. Nelson, and Robert A. Holt
- Subjects
Male ,Recurrence ,T-Lymphocytes ,Lymphoma, Non-Hodgkin ,Hematologic Neoplasms ,Immunology ,Antigens, CD19 ,Immunology and Allergy ,Humans ,Immunotherapy, Adoptive ,Cyclophosphamide ,Aged - Abstract
Access to commercial CD19 CAR-T cells remains limited even in wealthy countries like Canada due to clinical, logistical, and financial barriers related to centrally manufactured products. We created a non-commercial academic platform for end-to-end manufacturing of CAR-T cells within Canada’s publicly funded healthcare system. We report initial results from a single-arm, open-label study to determine the safety and efficacy of in-house manufactured CD19 CAR-T cells (entitled CLIC-1901) in participants with relapsed/refractory CD19 positive hematologic malignancies. Using a GMP compliant semi-automated, closed process on the Miltenyi Prodigy, T cells were transduced with lentiviral vector bearing a 4-1BB anti-CD19 CAR transgene and expanded. Participants underwent lymphodepletion with fludarabine and cyclophosphamide, followed by infusion of non-cryopreserved CAR-T cells. Thirty participants with non-Hodgkin’s lymphoma (n=25) or acute lymphoblastic leukemia (n=5) were infused with CLIC-1901: 21 males (70%), median age 66 (range 18-75). Time from enrollment to CLIC-1901 infusion was a median of 20 days (range 15-48). The median CLIC-1901 dose infused was 2.3 × 106 CAR-T cells/kg (range 0.13-3.6 × 106/kg). Toxicity included ≥ grade 3 cytokine release syndrome (n=2) and neurotoxicity (n=1). Median follow-up was 6.5 months. Overall response rate at day 28 was 76.7%. Median progression-free and overall survival was 6 months (95%CI 3-not estimable) and 11 months (95% 6.6-not estimable), respectively. This is the first trial of in-house manufactured CAR-T cells in Canada and demonstrates that administering fresh CLIC-1901 product is fast, safe, and efficacious. Our experience may provide helpful guidance for other jurisdictions seeking to create feasible and sustainable CAR-T cell programs in research-oriented yet resource-constrained settings.Clinical trial registrationhttps://clinicaltrials.gov/ct2/show/NCT03765177, identifier NCT03765177.
- Published
- 2022
6. Spontaneous remission and clonal evolution in lenalidomide associated secondary AML
- Author
-
Daniel D Waller, Yury Monczak, Michael Sebag, Chaim Shustik, and René P. Michel
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Remission, Spontaneous ,Spontaneous remission ,Newly diagnosed ,Secondary AML ,Somatic evolution in cancer ,Clonal Evolution ,03 medical and health sciences ,0302 clinical medicine ,Autologous stem-cell transplantation ,Internal medicine ,medicine ,Humans ,Lenalidomide ,Multiple myeloma ,business.industry ,Remission Induction ,Treatment options ,Hematology ,medicine.disease ,Leukemia, Myeloid, Acute ,030220 oncology & carcinogenesis ,Neoplasm Recurrence, Local ,business ,030215 immunology ,medicine.drug - Abstract
Lenalidomide maintenance following autologous stem cell transplantation (ASCT) has emerged as the preferred treatment option for newly diagnosed patients with multiple myeloma (MM). Two large rando...
- Published
- 2020
7. Single-cell transcriptional analysis of the immune tumour microenvironment during myeloma disease evolution
- Author
-
P. Leif Bergsagel, Zhihua Li, Danielle C Croucher, Daniel D Waller, Michael Sebag, Trevor J. Pugh, Xian Fang Huang, Laura M. Richards, Marta Chesi, and Suzanne Trudel
- Subjects
LAG3 ,Immune system ,medicine.anatomical_structure ,Tumor progression ,T cell ,medicine ,Cancer research ,Bone marrow ,Biology ,medicine.disease ,Multiple myeloma ,CD8 ,Natural killer cell - Abstract
Multiple myeloma is universally preceded by a premalignant disease state. However, efforts to develop preventative therapeutic strategies are hindered by an incomplete understanding of the immune mechanisms associated with progression. Using single-cell RNA-sequencing, we profiled 104,880 cells derived from the bone marrow of Vκ*MYC mice across the myeloma progression spectrum, of which 97,720 were identified as non-malignant cells of the tumour microenvironment. Analysis of the non-malignant cells comprising the immune microenvironment identified mechanisms associated with disease progression in innate and adaptive immune cell populations. This included activation of IL-17 signaling in myeloid cells from precursor mice, accompanied by upregulation of Il6 gene expression in basophils. In the T/Natural killer cell compartment, we identified Tox-expressing CD8+ T cells enriched in the tumour microenvironment of mice with overt disease, with co-expression of LAG3 and PD-1, as well as elevated T cell exhaustion signatures in mice with early disease. We subsequently showed that early intervention with combinatorial blockade of LAG3 and PD-1 using neutralizing monoclonal antibodies delayed tumor progression and improved survival of Vκ*MYC mice. Together, this work provides insight into the biology of myeloma evolution and nominates a treatment strategy for early disease.
- Published
- 2021
8. Oxo-aglaiastatin-Mediated Inhibition of Translation Initiation
- Author
-
Jennifer Chu, Rayelle Itoua Maïga, Daniel D Waller, Lauren E. Brown, William G. Devine, Jerry Pelletier, John A. Porco, Wenhan Zhang, Michael Sebag, and Regina Cencic
- Subjects
0301 basic medicine ,Lymphoma ,lcsh:Medicine ,Antineoplastic Agents ,Article ,03 medical and health sciences ,0302 clinical medicine ,Eukaryotic translation ,Cell Line, Tumor ,Neoplasms ,Eukaryotic initiation factor ,Animals ,Humans ,Peptide Chain Initiation, Translational ,lcsh:Science ,PI3K/AKT/mTOR pathway ,Sulfonamides ,Multidisciplinary ,Chemistry ,lcsh:R ,Drug Synergism ,Translation (biology) ,Biological activity ,EIF4A1 ,Bridged Bicyclo Compounds, Heterocyclic ,Antineoplastic Agents, Phytogenic ,RNA Helicase A ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Doxorubicin ,eIF4A ,Eukaryotic Initiation Factor-4A ,Female ,lcsh:Q ,Aglaia ,030217 neurology & neurosurgery - Abstract
Translation is a highly regulated process that is perturbed in human cancers, often through activation of the PI3K/mTOR pathway which impacts directly on the ribosome recruitment phase of translation initiation. While significant research has focused on “drugging” components of the PI3K/mTOR network, efforts have also been directed towards inhibiting eukaryotic initiation factor (eIF) 4F-dependent translation. Small molecule inhibitors of this complex have been identified, characterized, and used to validate the rationale of targeting this step to curtail tumor cell growth and modulate chemotherapy response. One such class of compounds are the rocaglates, secondary metabolites from the plant genus Aglaia, which target the RNA helicase subunit of eIF4F, eIF4A. Here we explore the ability of synthetic derivatives of aglaiastatins and an aglaroxin derivative to target the translation process in vitro and in vivo and find the synthetic derivative oxo-aglaiastatin to possess such activity. Oxo-aglaiastatin inhibited translation in vitro and in vivo and synergized with doxorubicin, ABT-199 (a Bcl-2 antagonist), and dexamethasone when tested on hematological cancer cells. The biological activity of oxo-aglaiastatin was shown to be a consequence of inhibiting eIF4A1 activity.
- Published
- 2019
9. Unraveling the Prenylation–Cancer Paradox in Multiple Myeloma with Novel Geranylgeranyl Pyrophosphate Synthase (GGPPS) Inhibitors
- Author
-
Félix Vincent, Mònica Gómez Palou, Jaeok Park, Michael Sebag, Albert M. Berghuis, Youla S. Tsantrizos, Viviane Ta, Cyrus M. Lacbay, Daniel D Waller, and Xian Fang Huang
- Subjects
Models, Molecular ,0301 basic medicine ,medicine.medical_treatment ,Protein Prenylation ,Apoptosis ,Inhibitory Concentration 50 ,03 medical and health sciences ,0302 clinical medicine ,Geranylgeranylation ,Downregulation and upregulation ,Prenylation ,Catalytic Domain ,Drug Discovery ,medicine ,Humans ,Enzyme Inhibitors ,Cell Proliferation ,Chemistry ,rap1 GTP-Binding Proteins ,Bisphosphonate ,3. Good health ,Pyrimidines ,030104 developmental biology ,Geranylgeranyl-Diphosphate Geranylgeranyltransferase ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Molecular Medicine ,Protein prenylation ,Multiple Myeloma ,Intracellular - Abstract
Post-translational prenylation of the small GTP-binding proteins (GTPases) is vital to a plethora of biological processes, including cellular proliferation. We have identified a new class of thienopyrimidine-based bisphosphonate (ThP-BP) inhibitors of the human geranylgeranyl pyrophosphate synthase (hGGPPS) that block protein prenylation in multiple myeloma (MM) cells leading to cellular apoptosis. These inhibitors are also effective in blocking the proliferation of other types of cancer cells. We confirmed intracellular target engagement, demonstrated the mechanism of action leading to apoptosis, and determined a direct correlation between apoptosis and intracellular inhibition of hGGPPS. Administration of a ThP-BP inhibitor to a MM mouse model confirmed in vivo downregulation of Rap1A geranylgeranylation and reduction of monoclonal immunoglobulins (M-protein, a biomarker of disease burden) in the serum. These results provide the first proof-of-principle that hGGPPS is a valuable therapeutic target in oncology and more specifically for the treatment of multiple myeloma.
- Published
- 2018
10. Inhibition of farnesyl pyrophosphate (FPP) and/or geranylgeranyl pyrophosphate (GGPP) biosynthesis and its implication in the treatment of cancers
- Author
-
Daniel D Waller, Youla S. Tsantrizos, and Jaeok Park
- Subjects
Models, Molecular ,Geranylgeranyl pyrophosphate ,Farnesyl pyrophosphate ,Protein Prenylation ,Mevalonic Acid ,Antineoplastic Agents ,GTPase ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Geranylgeranylation ,Prenylation ,Polyisoprenyl Phosphates ,Neoplasms ,Drug Discovery ,Animals ,Farnesyltranstransferase ,Humans ,Enzyme Inhibitors ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Chemistry ,030302 biochemistry & molecular biology ,Geranyltranstransferase ,Cell biology ,Biosynthetic Pathways ,Unfolded protein response ,Protein farnesylation ,Protein prenylation ,lipids (amino acids, peptides, and proteins) ,Sesquiterpenes - Abstract
Dysregulation of isoprenoid biosynthesis is implicated in numerous biochemical disorders that play a role in the onset and/or progression of age-related diseases, such as hypercholesterolemia, osteoporosis, various cancers, and neurodegeneration. The mevalonate metabolic pathway is responsible for the biosynthesis of the two key isoprenoid metabolites, farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP). Post-translational prenylation of various proteins, including the small GTP-binding proteins (GTPases), with either FPP or GGPP is vital for proper localization and activation of these proteins. Prenylated GTPases play a critical role in cell signaling, proliferation, cellular plasticity, oncogenesis, and cancer metastasis. Pre-clinical and clinical studies strongly suggest that inhibition of protein prenylation can be an effective treatment for non-skeletal cancers. In this review, we summarize the most recent drug discovery efforts focusing on blocking protein farnesylation and/or geranylgeranylation and the biochemical and structural data available in guiding the current on-going studies in drug discovery. Furthermore, we provide a summary on the biochemical association between disruption of protein prenylation, endoplasmic reticulum (ER) stress, unfolded protein response (UPR) signaling, and cancer.
- Published
- 2019
11. IL6 Promotes a STAT3-PRL3 Feedforward Loop via SHP2 Repression in Multiple Myeloma
- Author
-
Jing Yuan Chooi, Wee Joo Chng, Qi Zeng, Julia S.L. Lim, Daniel D Waller, Michael Sebag, Jianbiao Zhou, Tae-Hoon Chung, Yan Ting Hee, Phyllis S.Y. Chong, and Zea Tuan Tan
- Subjects
0301 basic medicine ,STAT3 Transcription Factor ,endocrine system ,Cancer Research ,Antineoplastic Agents ,Apoptosis ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Mice, SCID ,Biology ,Bortezomib ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,immune system diseases ,Mice, Inbred NOD ,hemic and lymphatic diseases ,medicine ,Tumor Cells, Cultured ,Gene silencing ,Animals ,Humans ,Phosphorylation ,Protein kinase B ,Multiple myeloma ,Cell Proliferation ,Regulation of gene expression ,Interleukin-6 ,medicine.disease ,Prognosis ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Survival Rate ,030104 developmental biology ,Oncology ,Proteasome ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Signal transduction ,Protein Tyrosine Phosphatases ,Multiple Myeloma ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug ,Signal Transduction - Abstract
Overexpression of PRL-3, an oncogenic phosphatase, was identified as a novel cluster in patients with newly diagnosed multiple myeloma. However, the regulation and oncogenic activities of PRL-3 in multiple myeloma warrant further investigation. Here, we report that IL6 activates STAT3, which acts as a direct transcriptional regulator of PRL-3. Upregulation of PRL-3 increased myeloma cell viability and rephosphorylated STAT3 in a biphasic manner through direct interaction and deactivation of SHP2, thus blocking the gp130 (Y759)-mediated repression of STAT3 activity. Abrogation of PRL-3 reduced myeloma cell survival, clonogenicity, and tumorigenesis, and detailed mechanistic studies revealed “deactivation” of effector proteins such as Akt, Erk1/2, Src, STAT1, and STAT3. Furthermore, loss of PRL-3 efficiently abolished nuclear localization of STAT3 and reduced its occupancy on the promoter of target genes c-Myc and Mcl-1, and antiapoptotic genes Bcl2 and Bcl-xL. PRL-3 also played a role in the acquired resistance of myeloma cells to bortezomib, which could be overcome by PRL-3 silencing. Of clinical relevance, STAT3 and PRL-3 expression was positively correlated in five independent cohorts, and the STAT3 activation signature was significantly enriched in patients with high PRL-3 expression. Furthermore, PRL-3 could be used as a biomarker to identify high-risk patients with multiple myeloma that exhibited poor prognosis and inferior outcome even when treated with novel combinational therapeutics (proteasome inhibitors and immunomodulatory imide drugs). Conclusively, our results support a feedforward mechanism between STAT3 and PRL-3 that prolongs prosurvival signaling in multiple myeloma, and suggest targeting PRL-3 as a valid therapeutic opportunity in multiple myeloma. Significance: IL6 promotes STAT3-dependent transcriptional upregulation of PRL-3, which in turn re-phosphorylates STAT3 and aberrantly activates STAT3 target genes, leading to bortezomib resistance in multiple myeloma.
- Published
- 2019
12. Natural history of relapsed myeloma, refractory to immunomodulatory drugs and proteasome inhibitors: a multicenter IMWG study
- Author
-
W. J. Chng, Michele Cavo, Shaji Kumar, Chang-Ki Min, Xavier Leleu, Charalampia Kyriakou, Sundar Jagannath, M.V. Mateos, Daniel D Waller, Je-Jung Lee, Ravi Vij, Evangelos Terpos, Chaim Shustik, Jens Hillengass, Heinz Ludwig, Dominik Dytfeld, Brian G.M. Durie, Parameswaran Hari, Hareth Nahi, Meral Beksac, Laura Rosiñol, Philippe Moreau, Ashraf Badros, Jin Seok Kim, Saad Z. Usmani, Ingela Turesson, Markus Hansson, Hyeon Seok Eom, H. Goldschmidt, Albert Oriol, E Kastritis, Tomer M Mark, Meletios A. Dimopoulos, Michel Delforge, J.J. Lahuerta, Elizabeth O'Donnell, J de la Rubia, Melissa Alsina, Paula Rodriguez Otero, Je-Hwan Lee, Ki-Hyun Kim, Ulf-Henrik Mellqvist, Henk M. Lokhorst, N W C J van de Donk, Shuji Ozaki, Enrique M. Ocio, Anna Sureda, Kumar, S. K, Dimopoulos, M. A, Kastritis, E, Terpos, E, Nahi, H, Goldschmidt, H, Hillengass, J, Leleu, X, Beksac, M, Alsina, M, Oriol, A, Cavo, M, Ocio, E. M, Mateos, M. V, O'Donnell, E. K, Vij, R, Lokhorst, H. M, van de Donk, N. W. C. J, Min, C, Mark, T, Turesson, I, Hansson, M, Ludwig, H, Jagannath, S, Delforge, M, Kyriakou, C, Hari, P, Mellqvist, U, Usmani, S. Z, Dytfeld, D, Badros, A. Z, Moreau, P, Kim, K, Otero, P. R, Lee, J. H, Shustik, C, Waller, D, Chng, W. J, Ozaki, S, Lee, J-J, de la Rubia, J, Eom, H. S, Rosinol, L, Lahuerta, J. J, Sureda, A, Kim, J. S, Durie, B. G. M., Hematology, and CCA - Cancer Treatment and quality of life
- Subjects
Oncology ,Male ,Cancer Research ,Improved survival ,Biochemistry ,Cohort Studies ,chemistry.chemical_compound ,Asia pacific ,0302 clinical medicine ,Recurrence ,Medicine ,Multiple myeloma ,Aged, 80 and over ,Treatment regimen ,Bortezomib ,Treatment options ,Hematology ,Middle Aged ,Prognosis ,Management ,Proteasome Inhibitor ,030220 oncology & carcinogenesis ,Female ,Multiple Myeloma ,Bristol-Myers ,Proteasome Inhibitors ,medicine.drug ,Human ,Adult ,medicine.medical_specialty ,Prognosi ,Immunology ,03 medical and health sciences ,Refractory ,Adjuvants, Immunologic ,Internal medicine ,Overall survival ,Humans ,In patient ,Survival analysis ,Lenalidomide ,Aged ,business.industry ,Cell Biology ,medicine.disease ,Pomalidomide ,Carfilzomib ,Survival Analysis ,Surgery ,Board (committee) ,Regimen ,chemistry ,Cohort Studie ,business ,030215 immunology - Abstract
Background: Treatment of multiple myeloma has evolved considerably in the past few years with availability of several news drugs as well as increasing use of multidrug combinations. These changes have no doubt led to the improved survival seen among patients with MM. We have previously shown that outcomes of patients intolerant or refractory to one of the IMiDs and bortezomib had a poor outcome. Since that time, other drugs of the same class as well as new classes of drugs have been introduced for the treatment of MM. We designed this retrospective study to estimate the outcomes in patients with relapsed myeloma, who have become refractory to the current generation IMiDs and proteasome inhibitors. Patients and Methods: Patients with relapsed multiple myeloma who have received at least 3 prior lines of therapy, is refractory to both an IMiD (lenalidomide or pomalidomide) AND a proteasome inhibitor (bortezomib or carfilzomib), and has been exposed to an alkylating agent were identified from multiple centers. The time patients met the above criteria was defined as T0, and details of all treatment regimens before and after T0 were collected using electronic CRFs. The study was approved by the IRB at the respective centers. Results: 543 patients were enrolled in this study; median age was 62 years (31-87) and 61% were males. Patients were enrolled from centers in North America (n=181), Europe (n=318), and Asia Pacific (n=44). Patients were diagnosed between 2006 and 2014, the median duration between diagnosis of myeloma and study entry (T0) was 3.1 years (0.3 to 9). The median (95% CI) estimated follow up from diagnosis and from T0 were 61 (57, 66) months and 13 (11, 15) months respectively. The median number of lines of therapy prior to T0 was 4 (3-13), 48% had a prior transplant. The median OS from T0 for the entire cohort was 13 (11, 15) months. For these 462 patients, the median number of recorded regimens was 2 (1-9). The overall response and the depth of response to each line of treatment following T0 are as shown in the table. The median (95% CI) PFS and OS from T0 was 5 (4, 6), and 15.2 (13, 17), respectively. The overall survival for the 81 patients with no treatment post T0 was only 2.1 months. In a multivariate analysis, duration from diagnosis to T0, ISS stage III and number of lines of therapy were all associated with inferior PFS, as well as OS, and in addition, serum creatinine>2 mg/dL at T0 also predicted inferior OS. Conclusions: The study provides the expected outcome following development of myeloma that is refractory to a PI and an IMiD. The outcomes of these patients appear to be better than we had seen historically in patients refractory/ intolerant to bortezomib and IMiDs, highlighting the increased treatment options available for these patients. However, there is decreasing response rate to sequential regimens highlighting the development of drug resistance. The data provides a bench mark for comparison of new therapies that are being evaluated in this disease. Table Table. Disclosures Dimopoulos: Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: 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; Genesis: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Kastritis:Takeda: Consultancy, Honoraria; Genesis: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Amgen: Consultancy, Honoraria. Terpos:BMS: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Other: Travel expenses, Research Funding; Celgene: Honoraria; Takeda: Consultancy, Honoraria; Genesis: Consultancy, Honoraria, Other: Travel expenses; Amgen: Consultancy, Honoraria, Other: Travel expenses, Research Funding; Novartis: Honoraria. Hillengass:Sanofi: Research Funding; Novartis: Research Funding; Amgen: Consultancy, Honoraria; BMS: Honoraria; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria. Leleu:TEVA: Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria; LeoPharma: Honoraria; Amgen: Honoraria; Bristol-Myers Squibb: Honoraria; Pierre Fabre: Honoraria; Celgene: Honoraria; Novartis: Honoraria; Takeda: Honoraria. Oriol:Janssen: Honoraria, Other: Expert board committee; Amgen: Honoraria, Other: Expert board committee. Cavo:Celgene: Consultancy, Honoraria; Millennium: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Janssen-Cilag: Consultancy, Honoraria; Amgen: Consultancy, Honoraria. Mateos:Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees. Vij:Shire: Consultancy; Takeda: Consultancy, Research Funding; Jazz: Consultancy; Karyopharma: Consultancy; Janssen: Consultancy; Novartis: Consultancy; Celgene: Consultancy; Bristol-Myers Squibb: Consultancy; Amgen: Consultancy, Research Funding. Lokhorst:Genmab: Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding. van de Donk:Amgen: Research Funding; Janssen: Research Funding; BMS: Research Funding; Celgene: Research Funding. Mark:Onyx: Research Funding, Speakers Bureau; Millenium: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau. Ludwig:Amgen: Research Funding, Speakers Bureau; Takeda: Research Funding, Speakers Bureau; BMS: Speakers Bureau; Janssen: Speakers Bureau. Jagannath:Novartis: Consultancy; Janssen: Consultancy; Bristol-Myers Squibb: Consultancy; Celgene: Consultancy; Merck: Consultancy. Usmani:Array: Research Funding; Britsol-Myers Squibb: Consultancy, Research Funding; Skyline: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; BioPharma: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Onyx: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Sanofi: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Amgen: Consultancy, Research Funding, Speakers Bureau; Millenium: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Speakers Bureau. Dytfeld:Janssen Poland: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Research Funding; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees. Moreau:Novartis: Honoraria; Amgen: Honoraria; Celgene: Honoraria; Bristol-Myers Squibb: Honoraria; Takeda: Honoraria; Janssen: Honoraria, Speakers Bureau. Lee:Amgen: Membership on an entity's Board of Directors or advisory committees. Shustik:Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Millenium: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees. de la Rubia:Celgene: Consultancy; Bristol Myers: Consultancy; Amgen,: Consultancy; Janssen: Consultancy. Durie:Takeda: Consultancy; Amgen: Consultancy; Janssen: Consultancy.
- Published
- 2017
13. Abstract 684: Inhibition of translation by aglaiastatins: Mechanism of action
- Author
-
Daniel D Waller, Michael Sebag, Lauren E. Brown, Mònica Gómez Palou, Jerry Pelletier, Regina Cencic, John A. Porco, Rayelle Itoua Maïga, and Jennifer Chu
- Subjects
Cancer Research ,Chemistry ,RNA ,RNA Helicase A ,Eukaryotic initiation factor 4F ,Oncology ,Mechanism of action ,eIF4A ,Cancer research ,medicine ,Protein biosynthesis ,medicine.symptom ,Cytotoxicity ,PI3K/AKT/mTOR pathway - Abstract
Background: Secondary metabolites from plants of the Aglaia genus consist of several classes of compounds, including the cyclopenta[b]benzopyrans, benzo[b]oxepines and cyclopenta[b]benzofurans (rocaglates). The best characterized of these is silvestrol; a rocaglate, which has been shown to target eukaryotic initiation factor 4A (eIF4A), the RNA helicase subunit of the eukaryotic initiation factor 4F (eIF4F) complex. The formation of this complex is regulated by the PI3K/mTOR and Ras-MAPK pathways. Hence, being at the nexus of important oncogenic pathways, eIF4F represents an attractive target for cancer therapy. Silvestrol and its analogs have demonstrated potent activity in human tumor cell lines and xenograft models. Some of the most responsive mRNAs are those encoding oncogenic proteins such as Myc and Mcl-1. This places this group of compounds as promising therapeutic agents against Myc-driven cancers. Purpose of the study: The purpose of this study is to characterize a sub-group of Aglaia secondary metabolites known as aglaiastatins, which are characterized by the presence of a pyrimidone subunit fused to the cyclopenta[b]benzofuran structure, resulting in a pentacyclic skeleton. Method: Using in vitro and in vivo assays, we assessed the potency of representative aglaiastatins towards inhibition of protein synthesis and cytotoxicity of tumor cells. Results: We showed that aglaiastatins induce a specific inhibition of cap-dependent translation by interfering with eIF4A's RNA binding activity, similar to rocaglates. This strong correlation in the mechanism of action was further demonstrated in an eIF4AF163L rocaglate-resistant cell line. Aglaiastatins also demonstrated single agent potency in vitro against a diverse panel of human lymphoma cell lines as well as primary patient samples. In vivo, the compound of interest was found to have a chemosensitization capability, by reversing chemoresistance to doxorubicin in a pre-clinical murine lymphoma model. Conclusion: Our results indicate that the aglaiastatins also target eIF4A, similarly to rocaglates. Their activity against rocaglate-resistant cells indicates a high similarity in drug target binding pattern. Moreover, the targeting of cap-dependent translation through the RNA helicase allows for the potent activity of the drug against several lymphoma lines, regardless of their mutational landscape, thus providing a potential therapeutic opportunity against difficult-to-treat hematological malignancies. Citation Format: Rayelle Itoua Maïga, Regina Cencic, Jennifer Chu, Lauren E. Brown, Daniel Dirck Waller, Mònica Gómez Palou, Michael Sebag, John A. Porco, Jerry Pelletier. Inhibition of translation by aglaiastatins: Mechanism of action [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 684.
- Published
- 2018
14. A Covalent Cysteine-Targeting Kinase Inhibitor of Ire1 Permits Allosteric Control of Endoribonuclease Activity
- Author
-
Youla S. Tsantrizos, Kurt Dejgaard, Lekha Sleno, David Y. Thomas, Daniel D Waller, René Roy, Tze Chieh Shiao, Gregor Jansen, Makan Golizeh, Michael Sebag, Chloe Martel-Lorion, and John Mancuso
- Subjects
0301 basic medicine ,Saccharomyces cerevisiae Proteins ,Endoribonuclease activity ,Allosteric regulation ,Molecular Sequence Data ,Pyrimidinones ,Saccharomyces cerevisiae ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,03 medical and health sciences ,Allosteric Regulation ,Endoribonucleases ,medicine ,Humans ,Amino Acid Sequence ,Cysteine ,Molecular Biology ,Protein Kinase Inhibitors ,Kinase ,Organic Chemistry ,Rational design ,Transmembrane protein ,030104 developmental biology ,Mechanism of action ,Unfolded protein response ,Biocatalysis ,Mutagenesis, Site-Directed ,Unfolded Protein Response ,Molecular Medicine ,medicine.symptom ,Sequence Alignment - Abstract
The unfolded protein response (UPR) initiated by the transmembrane kinase/ribonuclease Ire1 has been implicated in a variety of diseases. Ire1, with its unique position in the UPR, is an ideal target for the development of therapies; however, the identification of specific kinase inhibitors is challenging. Recently, the development of covalent inhibitors has gained great momentum because of the irreversible deactivation of the target. We identified and determined the mechanism of action of the Ire1-inhibitory compound UPRM8. MS analysis revealed that UPRM8 inhibition occurs by covalent adduct formation at a conserved cysteine at the regulatory DFG+2 position in the Ire1 kinase activation loop. Mutational analysis of the target cysteine residue identified both UPRM8-resistant and catalytically inactive Ire1 mutants. We describe a novel covalent inhibition mechanism of UPRM8, which can serve as a lead for the rational design and optimization of inhibitors of human Ire1.
- Published
- 2015
15. Decreasing Poly(ADP-Ribose) Polymerase Activity Restores ΔF508 CFTR Trafficking
- Author
-
Donglei Zhang, Jie Liao, Graeme W. Carlile, Pierre Lesimple, Heidi M. Sampson, Haouaria Balghi, Daniel D Waller, David Y. Thomas, Romeo Phillipe, Suzana M. Anjos, Françoise Dantzer, Julie Goepp, Pasquale Ferraro, Renaud Robert, Fabiana Ciciriello, and John W. Hanrahan
- Subjects
DF508 CFTR ,Transgene ,Cell ,Inflammation ,ABT-888 ,PARP-1 ,Biology ,Cystic fibrosis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,PARP1 ,medicine ,oxidative stress ,Pharmacology (medical) ,ΔF508 ,030304 developmental biology ,Original Research ,Pharmacology ,0303 health sciences ,Activator (genetics) ,lcsh:RM1-950 ,CF ,Sciences du Vivant [q-bio]/Biotechnologies ,16. Peace & justice ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,lcsh:Therapeutics. Pharmacology ,chemistry ,Immunology ,medicine.symptom ,DNA damage PARP-1−/− ,030217 neurology & neurosurgery ,Peroxynitrite - Abstract
Most cystic fibrosis is caused by mutations in CFTR that prevent its trafficking from the ER to the plasma membrane and is associated with exaggerated inflammation, altered metabolism, and diminished responses to oxidative stress. PARP-1 is activated by oxidative stress and causes energy depletion and cell dysfunction. Inhibition of this enzyme protects against excessive inflammation and recent studies have also implicated it in intracellular protein trafficking. We hypothesized that PARP-1 activity is altered in CF and affects trafficking and function of the most common CF mutant ΔF508 CFTR. Indeed, PARP-1 activity was 2.9-fold higher in CF (ΔF508/ΔF508) human bronchial epithelial primary cells than in non-CF cells, and similar results were obtained by comparing CF vs. non-CF bronchial epithelial cell lines (2.5-fold higher in CFBE41o(-) vs. 16HBE14o(-), P 0.002). A PARP-1 inhibitor (ABT-888, Veliparib) partially restored CFTR channel activity in CFBE41o(-) cells overexpressing ΔF508 CFTR. Similarly, reducing PARP-1 activity by 85% in ileum from transgenic CF mice (Cftr(tm1)Eur) partially rescued ΔF508 CFTR activity to 7% of wild type mouse levels, and similar correction (7.8%) was observed in vivo by measuring salivary secretion. Inhibiting PARP-1 with ABT-888 or siRNA partially restored ΔF508 CFTR trafficking in cell lines, and most ΔF508 CFTR was complex glycosylated when heterologously expressed in PARP-1(-/-) mouse embryonic fibroblasts. Finally, levels of the mature glycoform of CFTR were reduced by peroxynitrite, a strong activator of PARP-1. These results demonstrate that PARP-1 activity is increased in CF, and identify a novel pathway that could be targeted by proteostatic correctors of CFTR trafficking.
- Published
- 2012
16. Rho5p is involved in mediating the osmotic stress response in Saccharomyces cerevisiae, and its activity is regulated via Msi1p and Npr1p by phosphorylation and ubiquitination
- Author
-
Cunle Wu, David Y. Thomas, Daniel D Waller, Malcolm Whiteway, and Robert B. Annan
- Subjects
rho GTP-Binding Proteins ,Saccharomyces cerevisiae Proteins ,Osmotic shock ,GTPase-activating protein ,Saccharomyces cerevisiae ,Regulator ,GTPase ,Biology ,Microbiology ,Suppression, Genetic ,Ubiquitin ,Osmotic Pressure ,Gene Expression Regulation, Fungal ,Kinase activity ,Phosphorylation ,Molecular Biology ,GTPase-Activating Proteins ,Ubiquitination ,General Medicine ,Articles ,biology.organism_classification ,Cell biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Chromatin Assembly Factor-1 ,Biochemistry ,biology.protein ,Protein Kinases ,Protein Binding - Abstract
Small GTPases of the Rho family act as molecular switches, and modulation of the GTP-bound state of Rho proteins is a well-characterized means of regulating their signaling activity in vivo. In contrast, the regulation of Rho-type GTPases by posttranslational modifications is poorly understood. Here, we present evidence of the control of the Saccharomyces cerevisiae Rho-type GTPase Rho5p by phosphorylation and ubiquitination. Rho5p binds to Ste50p, and the expression of the activated RHO5 ( Q91H ) allele in an Δ ste50 strain is lethal under conditions of osmotic stress. An overexpression screen identified RGD2 and MSI1 as being high-copy suppressors of the osmotic sensitivity of this lethality. Rgd2p had been identified as being a possible Rho5p GTPase-activating protein based on an in vitro assay; this result supports its function as a regulator of Rho5p activity in vivo. MSI1 was previously identified as being a suppressor of hyperactive Ras/cyclic AMP signaling, where it antagonizes Npr1p kinase activity and promotes ubiquitination. Here, we show that Msi1p also acts via Npr1p to suppress activated Rho5p signaling. Rho5p is ubiquitinated, and its expression is lethal in a strain that is compromised for proteasome activity. These data identify Rho5p as being a target of Msi1p/Npr1p regulation and describe a regulatory circuit involving phosphorylation and ubiquitination.
- Published
- 2008
17. Nup53p is a target of two mitotic kinases, Cdk1p and Hrr25p
- Author
-
Axel Dienemann, Daniel D Waller, Richard W. Wozniak, David Y. Thomas, Taras Makhnevych, Malcolm Whiteway, and C. Patrick Lusk
- Subjects
Cytoplasm ,Saccharomyces cerevisiae Proteins ,isolation & purification ,enzymology ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Polo-like kinase ,Saccharomyces cerevisiae ,Biology ,Biochemistry ,Vitro ,Spindle pole body ,Phosphorylation cascade ,Structural Biology ,CDC2 Protein Kinase ,Genetics ,pharmaceutical ,Phosphorylation ,Molecular Biology ,Mitosis ,Alleles ,Glutathione Transferase ,Cyclin-dependent kinase 1 ,Casein Kinase I ,Protein ,Cell Cycle ,Proteins ,Cell Biology ,Yeast ,Cell biology ,In Vitro ,Nuclear Pore Complex Proteins ,Mutation ,Nucleoporin ,Casein kinase 1 ,metabolism ,Plasmids ,Protein Binding ,Subcellular Fractions - Abstract
Nuclear pore complexes (NPCs) form channels across the nuclear envelope and provide the sole sites of molecular exchange between the cytoplasm and nucleoplasm. The NPC is a target of a number of post-translational modifications, including phosphorylation, yet the functions of these modifications are ill defined. Here, we have investigated the mitotic specific phosphorylation of a yeast nucleoporin Nup53p. Two kinases were identified that phosphorylate Nup53p: the mitotic kinase Cdk1p/Cdc2p/Cdc28p and the casein kinase Hrr25p. Hrr25p was identified by screening 119 yeast kinases for their ability to phosphorylate Nup53p in vitro. Conditional alleles of Hrr25p support the conclusion that Hrr25p phosphorylates Nup53p in vivo. We further demonstrated using solution binding and affinity purification assays, that Hrr25p directly binds Nup53p in an interaction that is destabilized by the phosphorylation of Nup53p. Consistent with this observation, we observed that Hrr25p moves between distinct locations in the cell during the cell cycle including the nucleus, the cortex of the emerging bud and the spindle pole bodies. Cdk1p also contributes to Nup53p phosphorylation as specific inhibition of Cdk1p or mutation of Cdk1p consensus sites partially blocked its phosphorylation. The ability of nup53 alleles containing Cdk1p site mutations to complement synthetic defects of nup53 Delta nup170 Delta strains is linked to a function for Nup53p in the spindle assembly checkpoint.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.