34 results on '"Tian Y. Zhang"'
Search Results
2. Epstein–Barr virus-positive lymphoproliferative disorder manifesting as pulmonary disease in a patient with acute myeloid leukemia: a case report
- Author
-
Ritika Dutta, Susanna Y. Miao, Paul Phan, Sebastian Fernandez-Pol, Parveen Shiraz, Dora Ho, Gabriel N. Mannis, and Tian Y. Zhang
- Subjects
Acute myeloid leukemia ,Epstein–Barr virus ,Post-transplant lymphoproliferative disorder ,Hematopoietic stem cell transplant ,Case report ,Medicine - Abstract
Abstract Background Patients with lymphoproliferative disorders following hematopoietic stem cell transplant (HSCT) most commonly present with fever and lymphadenopathy within the first 5 months of transplant. Pulmonary post-transplant lymphoproliferative disorder (PTLD) is a particularly aggressive and rapidly progressive disease, with high morbidity and mortality. There are a very limited number of reported pulmonary PTLD cases following HSCT in patients with acute myeloid leukemia (AML). Early diagnosis and detection of pulmonary PTLD is critical given its high lethality. However, variable clinical presentations and nonspecific radiographic findings make pulmonary PTLD difficult to distinguish from other more common causes of pulmonary disease in AML patients. Case presentation Here, we describe a 68-year-old Caucasian man who presented for salvage induction therapy following relapse of his AML after a haploidentical allogeneic HSCT 10 months earlier. He developed recurrent fevers, dry cough, and hypoxemia, with chest computed tomography (CT) showing bibasilar consolidations and increased nodularity without increased lymphadenopathy. His symptoms initially improved with antibiotic and antifungal therapy, but his follow-up chest CT showed progression of disease despite symptomatic improvement. Epstein–Barr virus (EBV) was detected in his blood by polymerase chain reaction (PCR), and a lung biopsy revealed monomorphic PTLD with B cells positive for EBV. Unfortunately, the patient’s condition rapidly deteriorated, and he passed away prior to treatment initiation. Conclusions To our knowledge, this is the first reported case of an AML patient developing pulmonary PTLD relatively late in his post-transplant course in the setting of relapsed disease and salvage therapy. Pulmonary PTLD, a rare but highly lethal disorder, can imitate the symptoms and radiographic findings of pneumonia, a common diagnosis in immunocompromised AML patients. This case illustrates the importance of considering pulmonary PTLD in the differential diagnosis for pulmonary disease in AML patients with a history of HSCT, especially in the setting of progressive radiographic findings despite broad antibacterial and antifungal therapy. Further, our case demonstrates the importance of biopsy and uninterrupted EBV DNA monitoring in the definitive diagnosis of PTLD, given nonspecific symptomatology and radiographic findings.
- Published
- 2021
- Full Text
- View/download PDF
3. Emerging Molecularly Targeted Therapies in Castration Refractory Prostate Cancer
- Author
-
Jesal C. Patel, Benjamin L. Maughan, Archana M. Agarwal, Julia A. Batten, Tian Y. Zhang, and Neeraj Agarwal
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Androgen deprivation therapy (ADT) with medical or surgical castration is the mainstay of therapy in men with metastatic prostate cancer. However, despite initial responses, almost all men eventually develop castration refractory metastatic prostate cancer (CRPC) and die of their disease. Over the last decade, it has been recognized that despite the failure of ADT, most prostate cancers maintain some dependence on androgen and/or androgen receptor (AR) signaling for proliferation. Furthermore, androgen independent molecular pathways have been identified as drivers of continued progression of CRPC. Subsequently, drugs have been developed targeting these pathways, many of which have received regulatory approval. Agents such as abiraterone, enzalutamide, orteronel (TAK-700), and ARN-509 target androgen signaling. Sipuleucel-T, ipilimumab, and tasquinimod augment immune-mediated tumor killing. Agents targeting classic tumorogenesis pathways including vascular endothelial growth factor, hepatocyte growth factor, insulin like growth factor-1, tumor suppressor, and those which regulate apoptosis and cell cycles are currently being developed. This paper aims to focus on emerging molecular pathways underlying progression of CRPC, and the drugs targeting these pathways, which have recently been approved or have reached advanced stages of development in either phase II or phase III clinical trials.
- Published
- 2013
- Full Text
- View/download PDF
4. Disease Characterization and Response Prediction in Myeloma Patients Undergoing Conventional and Cellular Therapies from Circulating Tumor DNA
- Author
-
Hitomi Hosoya, Mia Carleton, Kailee L Tanaka, Brian Sworder, Vanna Hovanky, George E Duran, Tian Y. Zhang, Michael S. Khodadoust, David B. Miklos, Sally Arai, David Iberri, Michaela Liedtke, Surbhi Sidana, and David M. Kurtz
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
5. Data from KIT Signaling Governs Differential Sensitivity of Mature and Primitive CML Progenitors to Tyrosine Kinase Inhibitors
- Author
-
Michael W. Deininger, Brian J. Druker, Jorge E. Cortes, Paul W. Manley, Christopher A. Eide, Tian Y. Zhang, Anthony D. Pomicter, Jamshid S. Khorashad, Anna M. Eiring, Ira L. Kraft, Zhimin Gu, Thomas O'Hare, and Amie S. Corbin
- Abstract
Imatinib and other BCR-ABL1 inhibitors are effective therapies for chronic myelogenous leukemia (CML), but these inhibitors target additional kinases including KIT, raising the question of whether off-target effects contribute to clinical efficacy. On the basis of its involvement in CML pathogenesis, we hypothesized that KIT may govern responses of CML cells to imatinib. To test this, we assessed the growth of primary CML progenitor cells under conditions of sole BCR-ABL1, sole KIT, and dual BCR-ABL1/KIT inhibition. Sole BCR-ABL1 inhibition suppressed mature CML progenitor cells, but these effects were largely abolished by stem cell factor (SCF) and maximal suppression required dual BCR-ABL1/KIT inhibition. In contrast, KIT inhibition did not add to the effects of BCR-ABL1 inhibition in primitive progenitors, represented by CD34+38− cells. Long-term culture-initiating cell assays on murine stroma revealed profound depletion of primitive CML cells by sole BCR-ABL1 inhibition despite the presence of SCF, suggesting that primitive CML cells are unable to use SCF as a survival factor upon BCR-ABL1 inhibition. In CD34+38+ cells, SCF strongly induced pAKTS473 in a phosphoinositide 3-kinase (PI3K)–dependent manner, which was further enhanced by inhibition of BCR-ABL1 and associated with increased colony survival. In contrast, pAKTS473 levels remained low in CD34+38− cells cultured under the same conditions. Consistent with reduced response to SCF, KIT surface expression was significantly lower on CD34+38− compared with CD34+38+ CML cells, suggesting a possible mechanism for the differential effects of SCF on mature and primitive CML progenitor cells. Cancer Res; 73(18); 5775–86. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Methods and Figures and Table from KIT Signaling Governs Differential Sensitivity of Mature and Primitive CML Progenitors to Tyrosine Kinase Inhibitors
- Author
-
Michael W. Deininger, Brian J. Druker, Jorge E. Cortes, Paul W. Manley, Christopher A. Eide, Tian Y. Zhang, Anthony D. Pomicter, Jamshid S. Khorashad, Anna M. Eiring, Ira L. Kraft, Zhimin Gu, Thomas O'Hare, and Amie S. Corbin
- Abstract
PDF file, 421K, Supplementary Figure 1: Specificity of imatinib, dasatinib, PPY-A, BAW667, and a SCF-blocking antibody on Mo7ep210BCR-ABL1 cells. Supplementary Figure 2: Comparison of CFU-GM colony formation upon sole BCR-ABL1 inhibition (PPY-A), sole KIT inhibition or dual BCR-ABL1/KIT inhibition. Supplementary Figure 3: Inhibition of KIT in human cells through use of a lentiviral vector for simultaneous expression of shKIT and GFP . Supplementary Figure 4: Analysis of CML CFU-GM colony growth following removal of the individual cytokines. Supplementary Figure 5: BCR-ABL1 and KIT signaling influence activation of AKT associated with a reduction of Foxo3A. Supplementary Table 1. Activity profile of BAW667.
- Published
- 2023
- Full Text
- View/download PDF
7. Niche-directed therapy in acute myeloid leukemia: optimization of stem cell competition for niche occupancy
- Author
-
Tian Y. Zhang, Disha Dalela, Maxwell R Lloyd, Shyam A. Patel, and Amy Fan
- Subjects
Cancer Research ,medicine.medical_treatment ,Niche ,Disease ,Biology ,Malignancy ,Article ,Targeted therapy ,Mice ,Bone Marrow ,hemic and lymphatic diseases ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Stem Cell Niche ,Hematopoietic stem cell ,Myeloid leukemia ,Hematology ,Hematopoietic Stem Cells ,medicine.disease ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,Cell Competition ,Cancer research ,Bone marrow ,Stem cell - Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy of stem cell origin that contributes to significant morbidity and mortality. The long-term prognosis remains dismal given the high likelihood for primary refractory or relapsed disease. An essential component of relapse is resurgence from the bone marrow. To date, the murine hematopoietic stem cell (HSC) niche has been clearly defined, but the human HSC niche is less well understood. The design of niche-based targeted therapies for AML must account for which cellular subsets compete for stem cell occupancy within respective bone marrow microenvironments. In this review, we highlight the principles of stem cell niche biology and discuss translational insights into the AML microenvironment as of 2021. Optimization of competition for niche occupancy is important for the elimination of measurable residual disease (MRD). Some of these novel therapeutics are in the pharmacologic pipeline for AML and may be especially useful in the setting of MRD.
- Published
- 2021
- Full Text
- View/download PDF
8. Outcomes with Molecularly Targeted Agents as Salvage Therapy Following Frontline Venetoclax + Hypomethylating Agent in Adults with Acute Myeloid Leukemia: A Multicenter Retrospective Analysis
- Author
-
Vishesh Khanna, Tali Azenkot, Selina (Qiuying) Liu, Jason Gilbert, Edna Cheung, Kimberly Lau, Daniel A Pollyea, Elie Traer, Brian A Jonas, Tian Y Zhang, and Gabriel N Mannis
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2023
- Full Text
- View/download PDF
9. Abstract 5707: Tumor and immune determinants of response to anti-BCMA CAR T-cell therapy in multiple myeloma using cell-free DNA
- Author
-
Mia Carleton, Hitomi Hosoya, Kailee L. Tanaka, Brian Sworder, Vanna Hovanky, Bita Sahaf, Matthew J. Frank, George E. Duran, Tian Y. Zhang, Sally Arai, David Iberri, Michaela Liedtke, David B. Miklos, Michael S. Khodadoust, Surbhi Sidana, and David M. Kurtz
- Subjects
Cancer Research ,Oncology - Abstract
Background: Multiple myeloma (MM) is an incurable disease with a heterogenous clinical course and genomic landscape. Autologous anti-BCMA chimeric antigen receptor (CAR) T-cells are a promising new therapy, but determinants of response and resistance are not well known. Cell-free DNA (cfDNA) is a useful tool to study MM as it allows for repeated, non-invasive tumor assessment. We apply a novel method for simultaneously tracking tumor mutations and CAR T-cells from cfDNA using Cancer Personalized Profiling by Deep Sequencing (CAPP-Seq). Methods: We designed a 480kb CAPP-Seq hybrid capture panel to identify mutations, track tumor burden and minimal residual disease, and detect cfDNA derived from the CAR transgene (CAR-cfDNA) in patients receiving idecabtagene vicleucel (ide-cel). Flow cytometry (FC) for enumeration of CAR T-cells was performed from peripheral blood mononuclear cells (PBMCs) when available. Results: We profiled 153 biologic samples, including plasma, PBMCs, and bone marrow mononuclear cells, from 15 patients receiving ide-cel and 18 healthy controls. We observed a median of 84 SNVs (range 30-277) prior to therapy. Patients with prolonged responses (>90 days) had significantly lower circulating tumor DNA (ctDNA) at day 28 post-infusion than patients with early progression ( Conclusions: Cell-free DNA is a promising biomarker for mutational genotyping, disease monitoring, and tracking CAR T-cells in MM. The persistence of CAR-cfDNA has particular prognostic importance and novel strategies to increase CAR persistence should be explored. Citation Format: Mia Carleton, Hitomi Hosoya, Kailee L. Tanaka, Brian Sworder, Vanna Hovanky, Bita Sahaf, Matthew J. Frank, George E. Duran, Tian Y. Zhang, Sally Arai, David Iberri, Michaela Liedtke, David B. Miklos, Michael S. Khodadoust, Surbhi Sidana, David M. Kurtz. Tumor and immune determinants of response to anti-BCMA CAR T-cell therapy in multiple myeloma using cell-free DNA. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5707.
- Published
- 2023
- Full Text
- View/download PDF
10. Enasidenib drives human erythroid differentiation independently of isocitrate dehydrogenase 2
- Author
-
Anupama Narla, Melissa Stafford, Ravindra Majeti, Raymond Yin, Eric Gars, Daniel Thomas, Tian Y. Zhang, Yusuke Nakauchi, Satinder Kaur, Armon Azizi, Ritika Dutta, Thomas Köhnke, and Miles H. Linde
- Subjects
0301 basic medicine ,IDH1 ,Aminopyridines ,Protoporphyrins ,Enasidenib ,IDH2 ,03 medical and health sciences ,0302 clinical medicine ,Erythroid Cells ,hemic and lymphatic diseases ,Humans ,Medicine ,Progenitor cell ,Triazines ,business.industry ,Concise Communication ,Myeloid leukemia ,Cell Differentiation ,General Medicine ,Hematopoietic Stem Cells ,Isocitrate Dehydrogenase ,Haematopoiesis ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Erythropoiesis ,Stem cell ,business - Abstract
Cancer-related anemia is present in more than 60% of newly diagnosed cancer patients and is associated with substantial morbidity and high medical costs. Drugs that enhance erythropoiesis are urgently required to decrease transfusion rates and improve quality of life. Clinical studies have observed an unexpected improvement in hemoglobin and RBC transfusion-independence in patients with acute myeloid leukemia (AML) treated with the isocitrate dehydrogenase 2 (IDH2) mutant-specific inhibitor enasidenib, leading to improved quality of life without a reduction in AML disease burden. Here, we demonstrate that enasidenib enhanced human erythroid differentiation of hematopoietic progenitors. The phenomenon was not observed with other IDH1/2 inhibitors and occurred in IDH2-deficient CRISPR-engineered progenitors independently of D-2-hydroxyglutarate. The effect of enasidenib on hematopoietic progenitors was mediated by protoporphyrin accumulation, driving heme production and erythroid differentiation in committed CD71(+) progenitors rather than hematopoietic stem cells. Our results position enasidenib as a promising therapeutic agent for improvement of anemia and provide the basis for a clinical trial using enasidenib to decrease transfusion dependence in a wide array of clinical contexts.
- Published
- 2020
- Full Text
- View/download PDF
11. Circulating Tumor DNA for Disease Characterization and Response Prediction in Myeloma Patients Undergoing Chimeric Antigen Receptor T-Cell Therapy
- Author
-
Hitomi Hosoya, Mia Carleton, Kailee Tanaka, Brian Sworder, Vanna Hovanky, George E Duran, Tian Y Zhang, Michael Khodadoust, David B. Miklos, Sally Arai, David Iberri, Michaela Liedtke, Surbhi Sidana, and David Kurtz
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
- Full Text
- View/download PDF
12. Targeting LSCs: Peeling Back the Curtain on the Metabolic Complexities of AML
- Author
-
Ravindra Majeti and Tian Y. Zhang
- Subjects
0303 health sciences ,Myeloid ,Venetoclax ,Cell ,Cell Biology ,Biology ,medicine.disease ,03 medical and health sciences ,Leukemia ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.anatomical_structure ,Nicotinamide metabolism ,Bridged Bicyclo Compounds ,chemistry ,hemic and lymphatic diseases ,Genetics ,medicine ,Cancer research ,Molecular Medicine ,Stem cell ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Summary Most patients with AML succumb to chemoresistant leukemia stem cells (LSCs), which persist and reinitiate disease years after clinical remission. In this issue of Cell Stem Cell, Jones et al. (2020) identify a therapeutically targetable mechanism of resistance to venetoclax in relapsed and refractory AML LSCs mediated by nicotinamide metabolism.
- Published
- 2020
- Full Text
- View/download PDF
13. Improved outcomes of octogenarians and nonagenarians with acute myeloid leukemia in the era of novel therapies
- Author
-
Ritika Dutta, Mark Y. Jeng, Gabriel N. Mannis, Irena T. Tan, and Tian Y. Zhang
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,MEDLINE ,Myeloid leukemia ,Hematology ,business - Published
- 2020
- Full Text
- View/download PDF
14. Epstein-Barr virus-positive lymphoproliferative disorder manifesting as pulmonary disease in a patient with acute myeloid leukemia: a case report
- Author
-
Susanna Y. Miao, Gabriel N. Mannis, Ritika Dutta, Parveen Shiraz, Dora Y. Ho, Tian Y. Zhang, Paul Phan, and Sebastian Fernandez-Pol
- Subjects
Lung Diseases ,Male ,medicine.medical_specialty ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Salvage therapy ,Lymphoproliferative disorders ,lcsh:Medicine ,Case Report ,Lung biopsy ,Gastroenterology ,Post-transplant lymphoproliferative disorder ,Epstein–Barr virus ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,hemic and lymphatic diseases ,Biopsy ,medicine ,Humans ,Aged ,Acute myeloid leukemia ,medicine.diagnostic_test ,business.industry ,lcsh:R ,Hematopoietic Stem Cell Transplantation ,General Medicine ,medicine.disease ,Lymphoproliferative Disorders ,Pneumonia ,Leukemia, Myeloid, Acute ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Differential diagnosis ,Hematopoietic stem cell transplant ,business ,Progressive disease ,030215 immunology - Abstract
Background Patients with lymphoproliferative disorders following hematopoietic stem cell transplant (HSCT) most commonly present with fever and lymphadenopathy within the first 5 months of transplant. Pulmonary post-transplant lymphoproliferative disorder (PTLD) is a particularly aggressive and rapidly progressive disease, with high morbidity and mortality. There are a very limited number of reported pulmonary PTLD cases following HSCT in patients with acute myeloid leukemia (AML). Early diagnosis and detection of pulmonary PTLD is critical given its high lethality. However, variable clinical presentations and nonspecific radiographic findings make pulmonary PTLD difficult to distinguish from other more common causes of pulmonary disease in AML patients. Case presentation Here, we describe a 68-year-old Caucasian man who presented for salvage induction therapy following relapse of his AML after a haploidentical allogeneic HSCT 10 months earlier. He developed recurrent fevers, dry cough, and hypoxemia, with chest computed tomography (CT) showing bibasilar consolidations and increased nodularity without increased lymphadenopathy. His symptoms initially improved with antibiotic and antifungal therapy, but his follow-up chest CT showed progression of disease despite symptomatic improvement. Epstein–Barr virus (EBV) was detected in his blood by polymerase chain reaction (PCR), and a lung biopsy revealed monomorphic PTLD with B cells positive for EBV. Unfortunately, the patient’s condition rapidly deteriorated, and he passed away prior to treatment initiation. Conclusions To our knowledge, this is the first reported case of an AML patient developing pulmonary PTLD relatively late in his post-transplant course in the setting of relapsed disease and salvage therapy. Pulmonary PTLD, a rare but highly lethal disorder, can imitate the symptoms and radiographic findings of pneumonia, a common diagnosis in immunocompromised AML patients. This case illustrates the importance of considering pulmonary PTLD in the differential diagnosis for pulmonary disease in AML patients with a history of HSCT, especially in the setting of progressive radiographic findings despite broad antibacterial and antifungal therapy. Further, our case demonstrates the importance of biopsy and uninterrupted EBV DNA monitoring in the definitive diagnosis of PTLD, given nonspecific symptomatology and radiographic findings.
- Published
- 2020
15. Heterogeneous Definitions of Secondary Acute Myeloid Leukemia (AML) Yield Distinct Outcomes in Response to First-Line Treatment with Hypomethylating Agents (HMA) and Venetoclax (Ven)
- Author
-
Paul Phan, Raymond Yin, Gabriel N. Mannis, Irena Tan, Matthew Schwede, and Tian Y. Zhang
- Subjects
Oncology ,medicine.medical_specialty ,Yield (engineering) ,Venetoclax ,business.industry ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,First line treatment ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Ven ,Medicine ,Secondary Acute Myeloid Leukemia ,business - Abstract
Background: The combination of HMA and venetoclax is now standard of care for patients with AML who are not candidates for intensive chemotherapy. Elderly patients are more likely to have secondary AML (sAML), although the presence of an antecedent hematologic malignancy is often not apparent by history. Lindsley et al (Blood, 2015) showed that a somatic mutation in SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, or STAG2 is >95% specific for sAML and associated with worse outcomes. While outcomes with HMA/ven in patients meeting standard criteria for sAML have recently been reported (Pullarkat, ASCO 2021), we set out to conduct a real-world analysis of sAML patients receiving HMA/ven, including those with a secondary mutation profile (SMP) as described by Lindsley et al. We hypothesized that-when treated with HMA/ven-outcomes of patients with SMP may be most similar to those with de novo AML. Methods: Patients diagnosed with AML at Stanford Cancer Institute from 4/2017-3/2021 and treated with front-line HMA/ven were retrospectively reviewed. These included patients previously treated with HMA monotherapy for an antecedent hematologic malignancy and those who had previously received ≤ 3 cycles of HMA monotherapy for AML. Responses were classified per the modified International Working Group response criteria. Overall survival (OS) was assessed for all patients, and for patients who had a complete response (CR) or CR with incomplete hematologic recovery (CRi), duration of response (DoR) was also assessed. Statistical analyses were performed in R using the logrank test, with hazard ratios (HR) computed using the Cox proportional hazards model. For multivariate analyses, p-values for a specific variable were calculated using Cox proportional hazards regression. Results: 82 patients met criteria for inclusion; 78 had valid response assessments and 49 (62.8%) had achieved a CR or CRi at first response assessment. Median age was 72 years, with 3 patients younger than 60. 62 patients were male, median ECOG performance status (PS) was 1, median Charlson Comorbidity Index (CCI) was 6, median time to death or end of follow-up from the start of treatment was 366 days, and 58% of patients had adverse risk AML per ELN guidelines. Fig 1a demonstrates demographics for de novo, sAML (excluding SMP), and patients with SMP AML. 13 patients met criteria for AML-MRC, 23 patients had prior history of antecedent hematologic malignancy (18 with MDS or CMML, 5 with MDS/MPN overlap or MPN), 12 had tAML, and 20 patients possessed a SMP and did not meet criteria for the other three categories of sAML. 14 patients with de novo AML were characterized by the absence of any of the above factors. Patients with de novo AML were less likely to have adverse risk disease (29% vs. 64% in others) and had lower CCI scores (mean 5.1 vs. 6.2) but had no significant differences in age, gender, follow-up time, or PS. There was no statistically significant difference in rates of CR/CRi between the different subgroups or the different types of sAML; 69% of patients with de novo AML, 79% of SMP patients, and 57% of patients with other types of sAML achieved a CR or CRi. However, SMP patients had response durations and OS patterns similar to patients with de novo AML (Fig 1b and 1c), and when grouped with de novo patients, both DoR (HR = 3.5, p = 0.047, Fig 1d) and OS (HR = 2.1, p = 0.042, Fig 1e) were significantly longer than those of the sAML patients. Neither DoR nor OS were significantly longer when the SMP patients were grouped with sAML patients (respectively: HR = 3.3, p = 0.22, Fig 1f; HR = 1.5, p = 0.37, Fig 1g). In multivariate Cox proportional regression adjusting for age, ELN risk category, CCI, and PS, worse OS for sAML patients was maintained relative to the SMP and de novo patients (HR 2.9, p = 0.036), although the difference in DoR was no longer significant (HR 4.4, p= 0.10). Conclusions: Patients meeting standard definitions of sAML had worse outcomes than those with de novo AML when treated with HMA/ven in a retrospective, real-world analysis. Although a secondary mutation profile as described by Lindsley et al may be helpful in identifying patients with sAML, when treated with HMA/ven, patients with this profile have outcomes that align more closely with those of patients with de novo AML. Figure 1 Figure 1. Disclosures Mannis: Astex, Forty Seven Inc/Gilead, Glycomimetics, and Jazz Pharmaceuticals: Research Funding; AbbVie, Agios, Astellas Pharma, Bristol Myers Squibb, Genentech, MacroGenics, Pfizer, and Stemline: Consultancy.
- Published
- 2021
- Full Text
- View/download PDF
16. Abstract 1505: Reprogramming cancer into antigen presenting cells as a novel immunotherapy
- Author
-
Christopher Dove, Ravindra Majeti, Lindsay P. Miller, Melissa Stafford, Eric Gars, Thomas Köhnke, Payton L. Marshall, Robbie G. Majzner, Ian L. Linde, Miles H. Linde, Sarah F. Gurev, Paul Phan, Feifei Zhao, Amy Fan, and Tian Y. Zhang
- Subjects
Cancer Research ,Oncology ,business.industry ,medicine.medical_treatment ,Cancer research ,Medicine ,Cancer ,Immunotherapy ,business ,Antigen-presenting cell ,medicine.disease ,Reprogramming - Abstract
As a therapeutic modality, cancer vaccination leverages the well-characterized ability of antigen presenting cells (APCs) to take up and present tumor antigens in an effort to stimulate potent anti-cancer T cell responses. However, clinical successes with therapeutic cancer vaccination remains limited. Here, we present a novel cancer vaccination approach utilizing myeloid lineage reprogramming to directly convert cancer cells into tumor reprogramed-APCs (TR-APCs).In order to understand the therapeutic potential of TR-APCs, we generated syngeneic murine B-cell acute lymphoblastic leukemia models amenable to myeloid lineage reprogramming via ectopic expression of the myeloid lineage transcription factors PU.1 (Spi1) and C/EBPα (CEBPA). Upon enforced expression of these factors, the resulting TR-APCs acquire both myeloid phenotype and function, including the capacity for phagocytosis and antigen presentation. Crucially, TR-APCs can present endogenous self-derived tumor antigens directly encoded in their genome, without the need for phagocytosis and processing of adjacent tumor cells. In vitro, leukemia-derived TR-APCs express enhanced levels of antigen presentation machinery and co-stimulatory molecules, and potently stimulate tumor-specific CD4+ and CD8+ T cells. While in vivo TR-APC induction elicits only a modest extension to overall survival in immunodeficient hosts, generation of TR-APCs in immunocompetent syngeneic animals leads to complete leukemia eradication and protection from subsequent re-challenge. Further analysis of the in vivo immunological response to TR-APC induction revealed oligoclonal T cell expansion and establishment of cancer-specific immunological memory. Strikingly, use of a dual flank tumor model revealed that local TR-APC induction is sufficient to elicit systemic immunity capable of eradicating distant metastatic sites. We extended this treatment modality beyond hematologic malignancies, demonstrating that some solid tumors, including sarcomas and carcinomas, are amenable to myeloid-lineage reprogramming into TR-APCs, and contribute to increased overall survival. Finally, we demonstrate the clinical applicability of this approach by generating TR-APCs from primary human B cell acute lymphoblastic leukemia (B-ALL) specimens. Strikingly, primary B-ALL-derived TR-APCs stimulate autologous patient-derived T cells, demonstrating the clinical potential of TR-APCs to enhance antitumor immunity in patients. Thus, TR-APCs represent a novel cancer vaccination therapeutic strategy with broad implications for clinical immuno-oncology. Citation Format: Miles H. Linde, Sarah F. Gurev, Paul Phan, Feifei Zhao, Eric J. Gars, Melissa Stafford, Thomas Köhnke, Payton L. Marshall, Amy C. Fan, Christopher G. Dove, Ian L. Linde, Lindsay P. Miller, Robbie G. Majzner, Tian Yi Zhang, Ravindra Majeti. Reprogramming cancer into antigen presenting cells as a novel immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1505.
- Published
- 2021
- Full Text
- View/download PDF
17. Routine Use of Gemtuzumab Ozogamicin in 7+3-Based Inductions for All 'Non-Adverse' Risk AML
- Author
-
Abdullah Ladha, Michaela Liedtke, Lori Muffly, Gavin Hui, Steven Coutre, Caroline Berube, Gabriel N. Mannis, Edna Cheung, Tian Y. Zhang, and Jason Gotlib
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,medicine.drug_class ,Gemtuzumab ozogamicin ,Immunology ,Monoclonal antibody ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Calicheamicin ,medicine ,Humans ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,Gemtuzumab ,body regions ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Aminoglycosides ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,business ,030215 immunology ,Conjugate ,medicine.drug - Abstract
Introduction: The addition of gemtuzumab ozogamicin (GO) to 7+3 chemotherapy for newly diagnosed acute myeloid leukemia (AML) has been shown to significantly improve event-free survival (EFS) for cytogenetically favorable-risk AML, with marginal benefit for intermediate-risk AML, and no benefit for cytogenetically adverse-risk AML. Of note, with the exception of mutated FLT3-ITD, little is known about the impact of GO in ELN 2017-defined genotypically adverse-risk AML, and a recent randomized trial found no EFS benefit for 7+3+GO in patients (pts) with genotypically favorable-risk, NPM1-mutated AML. Since 2017, our institution incorporated GO into 7+3-based inductions for all "non-adverse" risk AML pts, as defined by wild-type FLT3 and no abnormalities on rapid FISH analysis for del(5q)/monosomy 5, del(7q)/monosomy 7, and del(20q). We report our experience treating all pts with "non-adverse" risk AML-as defined by this algorithm-with 7+3+GO. Methods: An institutional database was queried in order to identify all pts ≥18 years old who received 7+3-based chemotherapy for newly diagnosed AML between 2017 and 2020; pts who received the FDA-approved fractionated dose of GO were included in the analysis. Data collection included demographic variables, karyotype/FISH, targeted PCR analyses, and multigene NGS panels for AML-related mutations including, but not limited to, mutations in FLT3, NPM1, CEBPA, TP53, RUNX1, and ASXL1. Outcome data included response to induction, relapse, and death, as well as hematopoietic cell transplant (HCT) rates, conditioning regimens, and post-transplant complications. Results: Between January 2017 and July 2020, 96 pts received 7+3-based induction at our institution. Of these, 29 (30%) received 7+3 in combination with GO. Median age at diagnosis was 46 years (range 23-66), with 17 (59%) males. Sixteen (55%) pts had ELN favorable-risk AML (5 [31%] by cytogenetics and 11 [69%] by genotype), 6 (21%) pts had ELN intermediate-risk AML, and 7 (24%) pts had ELN adverse-risk AML (4 [57%] by cytogenetics and 3 [43%] by genotype). Median time from diagnosis to start of induction was 4 days (range 0-43). For cytogenetically adverse-risk pts, median time from diagnostic bone marrow biopsy to receipt of adverse karyotype results was 8 days (7-14). Median time from start of induction to receipt of multigene NGS results for all pts was 15 days (3-32). Overall, 22 (76%) pts achieved remission. All genotypically adverse-risk pts (1 with mutated TP53 and 2 with mutated RUNX1) were refractory to induction, while 3 of 4 (75%) cytogenetically adverse-risk pts (1 with t(6;9), 1 with monosomy 7, and 2 with 11q23 abnormalities) achieved remission. Eight of the 29 (28%) pts proceeded to HCT, including 4 adverse-risk pts. Of the adverse-risk pts, all received myeloablative conditioning prior to HCT and 3 (75%) developed veno-occlusive disease (VOD), with 2 (50%) requiring defibrotide therapy. In favorable/intermediate-risk pts, 4 (18%) proceeded to HCT (2 intermediate-risk pts in first remission and 2 favorable-risk pts in second remission). Of these, 2 (50%) received myeloablative conditioning and 1 (25%) developed VOD. At last follow-up, 23 of 29 pts (79%) remained alive, with a median overall survival not reached (range 1-29 months) and a median EFS of 20 months (9-31). The percentage of ELN favorable-, intermediate-, and adverse-risk pts who remained event-free at last follow-up was 75%, 33%, and 43%, respectively. Discussion: This single-center, retrospective cohort describes the outcomes of pts with "non-adverse" risk AML who received induction chemotherapy with 7+3+GO according to a pre-defined algorithm. Using this algorithm, 30% of all pts receiving 7+3-based inductions received GO. Of these, nearly 25% were ultimately found to have adverse-risk AML as defined by ELN 2017 criteria, largely driven by long turn-around times for karyotyping and NGS multigene panel results. No patient with genotypically adverse-risk AML by ELN criteria responded to induction chemotherapy, and 75% of cytogenetically adverse-risk pts who proceeded to HCT developed VOD. Routine use of 7+3+GO induction outside of the context of cytogenetically favorable-risk AML remains controversial, and further study is needed to define the role of GO, particularly for pts with ELN genotypically adverse-risk AML. Table Disclosures Gotlib: Blueprint Medicines Corporation: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Chair of the Response Adjudication Committee and Research Funding, Research Funding; Deciphera: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: co-chair of the Study Steering Committee and Research Funding. Liedtke:Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Pfizer: Honoraria; GSK: Membership on an entity's Board of Directors or advisory committees; Adaptive: Membership on an entity's Board of Directors or advisory committees; Caelum: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Muffly:Adaptive: Research Funding; Amgen: Consultancy; Servier: Research Funding. Mannis:AbbVie, Agios, Bristol-Myers Squibb, Genentech: Consultancy; Glycomimetics, Forty Seven, Inc, Jazz Pharmaceuticals: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
18. IL-6 blockade reverses bone marrow failure induced by human acute myeloid leukemia
- Author
-
Ravindra Majeti, Raymond Yin, Brooks Benard, Tian Y. Zhang, Feifei Zhao, and Ritika Dutta
- Subjects
0301 basic medicine ,Anemia ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Blocking antibody ,medicine ,Animals ,Humans ,business.industry ,Interleukin-6 ,Bone marrow failure ,Myeloid leukemia ,Cell Differentiation ,General Medicine ,Bone Marrow Failure Disorders ,medicine.disease ,Blockade ,Transplantation ,Haematopoiesis ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Bone marrow ,business - Abstract
Most patients with acute myeloid leukemia (AML) die from complications arising from cytopenias resulting from bone marrow (BM) failure. The common presumption among physicians is that AML-induced BM failure is primarily due to overcrowding, yet BM failure is observed even with low burden of disease. Here, we use large clinical data sets to show the lack of correlation between BM blast burden and degree of cytopenias at the time of diagnosis. We develop a splenectomized xenograft model to demonstrate that transplantation of human primary AML into immunocompromised mice recapitulates the human disease course by induction of BM failure via depletion of mouse hematopoietic stem and progenitor populations. Using unbiased approaches, we show that AML-elaborated IL-6 acts to block erythroid differentiation at the proerythroblast stage and that blocking antibodies against human IL-6 can improve AML-induced anemia and prolong overall survival, suggesting a potential therapeutic approach.
- Published
- 2019
19. NTIRE 2019 challenge on video super-resolution: methods and results
- Author
-
Tekalp, Ahmet Murat (ORCID 0000-0003-1465-8121 & YÖK ID 26207), Nah, S.; Timofte, R.; Gu, S.; Baik, S.; Hong, S.; Moon, G.; Son, S.; Lee, K.M.; Wang, X.; Chan, K.C.K.; Yu, K.; Dong, C.; Loy, C.C.; Fan, Y.; Yu, J.; Liu, D.; Huang, T.S.; Liu, X.; Li, C.; He, D.; DIng, Y.; Wen, S.; Porikli, F.; Kalarot, R.; Haris, M.; Shakhnarovich, G.; Ukita, N.; Yi, P.; Wang, Z.; Jiang, K.; Jiang, J.; Ma, J.; Dong, H.; Zhang, X.; Hu, Z.; Kim, K.; Kang, D.U.; Chun, S.Y.; Purohit, K.; Rajagopalan, A.N.; Tian, Y.; Zhang, Y.; Fu, Y.; Xu, C.; Yılmaz, M.A.; Korkmaz, C.; Sharma, M.; Makwana, M.; Badhwar, A.; Singh, A.P.; Upadhyay, A.; Mukhopadhyay, R.; Shukla, A.; Khanna, D.; Mandal, A.S.; Chaudhury, S.; Miao, S.; Zhu, Y.; Huo, X., College of Engineering, Department of Electrical and Electronics Engineering, Tekalp, Ahmet Murat (ORCID 0000-0003-1465-8121 & YÖK ID 26207), Nah, S.; Timofte, R.; Gu, S.; Baik, S.; Hong, S.; Moon, G.; Son, S.; Lee, K.M.; Wang, X.; Chan, K.C.K.; Yu, K.; Dong, C.; Loy, C.C.; Fan, Y.; Yu, J.; Liu, D.; Huang, T.S.; Liu, X.; Li, C.; He, D.; DIng, Y.; Wen, S.; Porikli, F.; Kalarot, R.; Haris, M.; Shakhnarovich, G.; Ukita, N.; Yi, P.; Wang, Z.; Jiang, K.; Jiang, J.; Ma, J.; Dong, H.; Zhang, X.; Hu, Z.; Kim, K.; Kang, D.U.; Chun, S.Y.; Purohit, K.; Rajagopalan, A.N.; Tian, Y.; Zhang, Y.; Fu, Y.; Xu, C.; Yılmaz, M.A.; Korkmaz, C.; Sharma, M.; Makwana, M.; Badhwar, A.; Singh, A.P.; Upadhyay, A.; Mukhopadhyay, R.; Shukla, A.; Khanna, D.; Mandal, A.S.; Chaudhury, S.; Miao, S.; Zhu, Y.; Huo, X., College of Engineering, and Department of Electrical and Electronics Engineering
- Abstract
This paper reviews the first NTIRE challenge on video super-resolution (restoration of rich details in low-resolution video frames) with focus on proposed solutions and results. A new REalistic and Diverse Scenes dataset (REDS) was employed. The challenge was divided into 2 tracks. Track 1 employed standard bicubic downscaling setup while Track 2 had realistic dynamic motion blurs. Each competition had 124 and 104 registered participants. There were total 14 teams in the final testing phase. They gauge the state-of-the-art in video super-resolution., NA
- Published
- 2019
20. Over-expression of TRPM8 is associated with poor prognosis in urothelial carcinoma of bladder
- Author
-
Xing Zhou, Lei M. Jiang, Xiao K. Zhao, Bo Ge, Tian Y. Zhang, and Ning Xiao
- Subjects
Male ,Oncology ,Pathology ,medicine.medical_specialty ,Urinary Bladder ,TRPM Cation Channels ,Biology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Immunoenzyme Techniques ,fluids and secretions ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,RNA, Messenger ,Survival rate ,Survival analysis ,Aged ,Neoplasm Staging ,Reverse Transcriptase Polymerase Chain Reaction ,Proportional hazards model ,Case-control study ,General Medicine ,Prognosis ,Survival Rate ,Reverse transcription polymerase chain reaction ,Real-time polymerase chain reaction ,Urinary Bladder Neoplasms ,Case-Control Studies ,embryonic structures ,Immunohistochemistry ,Female ,Neoplasm Grading ,Carcinogenesis ,Follow-Up Studies - Abstract
Transient receptor protein (TRP) channels are frequently associated with tumors and are correlated to patient’s outcome. We firstly investigated TRP channel melastatin 8 (TRPM8) expression in urothelial carcinoma of bladder (UCB) and its correlation with UCB clinicopathological features and additionally evaluated the association between TRPM8 expression and patients’ survival rate to elucidate its role in bladder oncogenesis. Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was conducted to examine TRPM8 messenger RNA (mRNA) expression in 36 pairs of freshly frozen UCB tissues and matched noncancerous tissues. Immunohistochemistry (IHC) was performed in 156 archived paraffin-embedded UCB samples to explore the correlation between TRPM8 protein and clinicopathological features. The association between TRPM8 expression and patient’s survival rate was evaluated using the Kaplan–Meier method and multivariate Cox regression analysis, respectively. The expression levels of TRPM8 mRNA in UCB tissues were significantly higher than those in matched noncancerous tissues (P = 0.016). Expression of TRPM8 protein in UCB was significantly and positively associated with histological grade (P = 0.039) and tumor stage (P = 0.037). Significant correlation between high TRPM8 expression and poor cumulative survival of UCB patients was shown using the Kaplan–Meier survival curve (P = 0.039). TRPM8 was represented as an independent prognostic biomarker for UCB patients by multivariate Cox regression analysis (P = 0.047). The present study provide the convincing evidence for the first time that over-expression of TRPM8 may play a role in the pathogenesis and progression of UCB, and TRPM8 may serve as an independent prognostic biomarker for UCB patients.
- Published
- 2014
- Full Text
- View/download PDF
21. Enasidenib Drives Maturation of Human Erythroid Precursors Independently of IDH2
- Author
-
Satinder Kaur, Ritika Dutta, Anupama Narla, Thomas Koehnke, Melissa Stafford, Tian Y. Zhang, Daniel Thomas, Ravindra Majeti, Raymond Yin, Eric Gars, and Yusuke Nakauchi
- Subjects
Chemistry ,Immunology ,Transferrin receptor ,Cell Biology ,Hematology ,Enasidenib ,Mitochondrion ,Biochemistry ,IDH2 ,Cell biology ,Precursor cell ,Idh2 gene ,Stem cell ,Interleukin 3 - Abstract
Acute Myeloid Leukemia (AML) remains one of the most difficult cancers to treat, with a 30% 2-year survival rate. High-throughput sequencing of AML patients has identified mutations, including FLT3, IDH1, and IDH2, for which targeted therapies have been developed. Enasidenib is an FDA-approved, first-in-class agent that preferentially inhibits IDH2-mutant activity and reduces levels of the oncometabolite 2-HG, inducing differentiation of IDH2-mutated blasts. Interestingly, greater than 50% of enasidenib-treated patients who had no objective clinical response still demonstrated improvement in their peripheral blood counts and reached RBC transfusion independence. The mechanism underlying this phenomenon is unknown but is of great clinical relevance given the high transfusion dependence and anemia-associated complications universally associated with AML. Thus, we sought to investigate how enasidenib drives normal hematopoiesis to improve quality of life and reduce morbidity in AML patients. In this study, we demonstrate that enasidenib enhances erythropoiesis from normal CD34+ hematopoietic stem and progenitor cells (HSPCs) derived from cord blood (CB) and bone marrow. Enasidenib doubled the proportion and total number of mature CD71+/GPA+ erythroblasts after 8 days of culture with EPO, SCF, and IL-3. In the presence of EPO, enasidenib induced a gene signature characteristic of maturing erythrocytes, with increased expression of GATA1 (1.3 fold), EPOR (2 fold), and KLF1 (1.4 fold), and decreased PU.1 (0.5 fold) and GATA2 (0.7 fold). Enasidenib-treated progenitor cells further demonstrated increased hemoglobin production (1.9 fold) and morphologic characteristics of increased erythroid maturation. Next, we sought to determine if enasidenib augments erythroid differentiation through IDH2 and IDH2-dependent pathways. First, we found that other IDH inhibitors (AG-120, AGI-6780, and AG-881) did not increase erythropoiesis at doses ranging from 1-10μM. As expected for normal HSPCs, 2-HG was not present at detectable levels in either the DMSO or enasidenib-treated conditions, and addition of 2-HG (50, 200μM) did not affect the ability of enasidenib to increase the proportion of CD71+GPA+ cells. Because it is possible that enasidenib acts through inhibition of wild-type IDH2, we generated CRISPR-Cas9 engineered IDH2 knockout (KO) CD34+ cells and treated them with enasidenib. Similar to wildtype cells, IDH2 KO CB CD34+ cells demonstrated a 3.4-fold increase in %CD71+GPA+ erythroid cells. Thus, enasidenib augments erythropoiesis independently of both mutant and wildtype IDH2 pathways. We then investigated the progenitor population that enasidenib acts on to drive erythroid maturation. Enasidenib did not increase the number of BFU-E or CFU-E colonies or the proportion of BFU-E (IL3R-CD34+CD36-) and CFU-E (IL3R-CD34-CD36+) progenitors in colony forming or liquid culture assays, respectively, leading us to conclude that enasidenib acts on more mature erythroid progenitors. Indeed, treating sorted mature CD71+ erythroid progenitors with enasidenib increased %CD71+GPA+ cells compared to DMSO control, whereas enasidenib treatment of CD71- early erythroid progenitors showed no effect. These observations provide evidence that enasidenib acts on CD71+ erythroid progenitors to increase late-stage erythroid differentiation. Given that CD71 allows for iron uptake into erythroid progenitors, we hypothesized that enasidenib modulates the heme biosynthesis pathway. Enasidenib inhibited the ABCG2 transporter, which effluxes protoporphyrin IX (PPIX), the direct precursor to heme, from the mitochondria and cytosol. Inhibition of ABCG2 by enasidenib could lead to PPIX accumulation within the cell, driving increased heme synthesis. To investigate this hypothesis, we treated cells with 20μM Ko143, a potent ABCG2 inhibitor, and observed a similar increase in %CD71+GPA+ cells as seen with enasidenib. Measurement of PPIX autofluorescence by flow cytometry and microscopy revealed an increase of PPIX in enasidenib-treated cells by 1.2-fold. Together, our data suggests that enasidenib drives maturation of CD71+ erythroid precursors independently of wildtype or mutant IDH2. Our results position enasidenib as a promising therapy to stimulate erythropoiesis and provide the basis for a clinical trial using enasidenib to improve anemia in a wide array of clinical contexts. Disclosures Majeti: Forty Seven Inc.: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties; BioMarin: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
22. KIT Signaling Governs Differential Sensitivity of Mature and Primitive CML Progenitors to Tyrosine Kinase Inhibitors
- Author
-
Jamshid S. Khorashad, Christopher A. Eide, Amie S. Corbin, Tian Y. Zhang, Ira L. Kraft, Brian J. Druker, Anna M. Eiring, Paul W. Manley, Michael W. Deininger, Zhimin Gu, Anthony D. Pomicter, Thomas O'Hare, and Jorge E. Cortes
- Subjects
Cancer Research ,Stromal cell ,Cellular differentiation ,Blotting, Western ,Fusion Proteins, bcr-abl ,CD34 ,Fluorescent Antibody Technique ,Antigens, CD34 ,Apoptosis ,Stem cell factor ,Biology ,Real-Time Polymerase Chain Reaction ,Piperazines ,Article ,Mice ,Phosphatidylinositol 3-Kinases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,RNA, Messenger ,Progenitor cell ,Protein Kinase Inhibitors ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Stem Cell Factor ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Differentiation ,Imatinib ,Flow Cytometry ,medicine.disease ,Molecular biology ,Proto-Oncogene Proteins c-kit ,Pyrimidines ,Oncology ,Drug Resistance, Neoplasm ,Benzamides ,Imatinib Mesylate ,Neoplastic Stem Cells ,Cancer research ,Stromal Cells ,Tyrosine kinase ,medicine.drug ,Chronic myelogenous leukemia - Abstract
Imatinib and other BCR-ABL1 inhibitors are effective therapies for chronic myelogenous leukemia (CML), but these inhibitors target additional kinases including KIT, raising the question of whether off-target effects contribute to clinical efficacy. On the basis of its involvement in CML pathogenesis, we hypothesized that KIT may govern responses of CML cells to imatinib. To test this, we assessed the growth of primary CML progenitor cells under conditions of sole BCR-ABL1, sole KIT, and dual BCR-ABL1/KIT inhibition. Sole BCR-ABL1 inhibition suppressed mature CML progenitor cells, but these effects were largely abolished by stem cell factor (SCF) and maximal suppression required dual BCR-ABL1/KIT inhibition. In contrast, KIT inhibition did not add to the effects of BCR-ABL1 inhibition in primitive progenitors, represented by CD34+38− cells. Long-term culture-initiating cell assays on murine stroma revealed profound depletion of primitive CML cells by sole BCR-ABL1 inhibition despite the presence of SCF, suggesting that primitive CML cells are unable to use SCF as a survival factor upon BCR-ABL1 inhibition. In CD34+38+ cells, SCF strongly induced pAKTS473 in a phosphoinositide 3-kinase (PI3K)–dependent manner, which was further enhanced by inhibition of BCR-ABL1 and associated with increased colony survival. In contrast, pAKTS473 levels remained low in CD34+38− cells cultured under the same conditions. Consistent with reduced response to SCF, KIT surface expression was significantly lower on CD34+38− compared with CD34+38+ CML cells, suggesting a possible mechanism for the differential effects of SCF on mature and primitive CML progenitor cells. Cancer Res; 73(18); 5775–86. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
23. Erratum: Combined STAT3 and BCR-ABL1 inhibition induces synthetic lethality in therapy-resistant chronic myeloid leukemia
- Author
-
Derek J. Wilson, Riccardo Baron, Ira L. Kraft, Matthew S. Zabriskie, William L. Heaton, Michael W. Deininger, Anna M. Eiring, Anna V. Senina, Brent D. G. Page, Nadeem A. Vellore, Thomas O'Hare, Robert Colaguori, Carolynn C. Arpin, Patrick T. Gunning, Tian Y. Zhang, S Ahmad, Diana Resetca, Kimberly R. Reynolds, Richard Moriggl, Jamshid S. Khorashad, Clinton C. Mason, A Todic, Srinivas K. Tantravahi, A J Engar, David J. Anderson, and Anthony D. Pomicter
- Subjects
Cancer Research ,Dasatinib ,Fusion Proteins, bcr-abl ,Apoptosis ,Synthetic lethality ,Tyrosine-kinase inhibitor ,Piperazines ,0302 clinical medicine ,Genes, Reporter ,hemic and lymphatic diseases ,Drug Discovery ,Phosphorylation ,Luciferases ,0303 health sciences ,Sulfonamides ,Gene Expression Regulation, Leukemic ,Myeloid leukemia ,Hematology ,3. Good health ,Molecular Docking Simulation ,Leukemia ,Haematopoiesis ,Oncology ,030220 oncology & carcinogenesis ,Benzamides ,Imatinib Mesylate ,Neoplastic Stem Cells ,medicine.drug ,Signal Transduction ,STAT3 Transcription Factor ,medicine.drug_class ,Antineoplastic Agents ,Biology ,Article ,Small Molecule Libraries ,03 medical and health sciences ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,Kinase activity ,Protein Kinase Inhibitors ,030304 developmental biology ,medicine.disease ,respiratory tract diseases ,Protein Structure, Tertiary ,Aminosalicylic Acids ,Thiazoles ,Imatinib mesylate ,Pyrimidines ,Drug Resistance, Neoplasm ,Cancer research ,Leukocytes, Mononuclear - Abstract
Mutations in the BCR-ABL1 kinase domain are an established mechanism of tyrosine kinase inhibitor (TKI) resistance in Philadelphia chromosome-positive leukemia, but fail to explain many cases of clinical TKI failure. In contrast, it is largely unknown why some patients fail TKI therapy despite continued suppression of BCR-ABL1 kinase activity, a situation termed BCR-ABL1 kinase-independent TKI resistance. Here, we identified activation of signal transducer and activator of transcription 3 (STAT3) by extrinsic or intrinsic mechanisms as an essential feature of BCR-ABL1 kinase-independent TKI resistance. By combining synthetic chemistry, in vitro reporter assays, and molecular dynamics-guided rational inhibitor design and high-throughput screening, we discovered BP-5-087, a potent and selective STAT3 SH2 domain inhibitor that reduces STAT3 phosphorylation and nuclear transactivation. Computational simulations, fluorescence polarization assays and hydrogen–deuterium exchange assays establish direct engagement of STAT3 by BP-5-087 and provide a high-resolution view of the STAT3 SH2 domain/BP-5-087 interface. In primary cells from chronic myeloid leukemia (CML) patients with BCR-ABL1 kinase-independent TKI resistance, BP-5-087 (1.0 μM) restored TKI sensitivity to therapy-resistant CML progenitor cells, including leukemic stem cells. Our findings implicate STAT3 as a critical signaling node in BCR-ABL1 kinase-independent TKI resistance, and suggest that BP-5-087 has clinical utility for treating malignancies characterized by STAT3 activation.
- Published
- 2017
- Full Text
- View/download PDF
24. Human Acute Myeloid Leukemia Inhibits Normal Erythroid Differentiation through the Paracrine Effects of IL-6
- Author
-
Ravindra Majeti, Feifei Zhao, Tian Y. Zhang, and Ritika Dutta
- Subjects
Myeloid ,business.industry ,Immunology ,CD34 ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Extramedullary hematopoiesis ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Cancer research ,Medicine ,Erythropoiesis ,Bone marrow ,business - Abstract
Acute Myeloid Leukemia (AML) is an aggressive cancer resulting in severe cytopenias related to bone marrow (BM) failure. The common assumption for AML-induced BM failure is overcrowding due to clonal expansion of immature myeloid blasts, leading to failure of normal hematopoiesis. However, in a cohort of 293 AML patients, we found that disease burden (% of blasts determined on diagnostic BM aspirate) did not predict severity of cytopenia (hemoglobin rs=-0.053; p=0.49; WBC rs=-0.030, p=0.70; platelet rs=0.091, p=0.026), strongly arguing against simple crowding as the main mechanism underlying AML-induced BM failure. Thus, the goal of our study is to identify novel mechanism(s) associated with AML-induced BM failure, potentially enabling development of new therapies to improve AML management and reverse morbidity. Conventional xenograft models of human AML do not typically exhibit cytopenias, making them unsuitable to study AML-induced BM failure. We speculated that in the mouse, increased splenic extramedullary hematopoiesis compensated for failed intramedullary hematopoiesis due to AML. To test this hypothesis, we performed surgical splenectomy on NSG mice prior to their transplantation with human AML. Strikingly, splenectomized NSG mice engrafted with primary human AML at a 30-70% disease burden (n=8 for primary AML samples, n=5-10 for each group of splenectomized NSG mice) developed leukopenia and severe anemia compared to sham-operated AML-engrafted controls. AML-engrafted splenectomized NSG mice showed early mortality compared to AML-engrafted NSG mice with intact spleens (10.1 weeks vs. 34.2 weeks p Utilizing our model, splenectomized NSG mice engrafted with human AML demonstrated depletion of normal hematopoietic progenitors (HSPCs) including HSCs (11.1 fold p To explore mechanisms by which AML blasts inhibit normal HSPCs, we generated conditioned media (CM) from patient-derived AML blasts, and found that AML-CM suppressed BFU-E colony formation from normal HSPCs (3.1-5.1 fold). Furthermore, in a direct co-culture system with AML blasts and CD34+ HSPCs, AML blasts inhibited erythroid differentiation from the CFU-E to normoblast stage by 81%. Removing CD34+ cells from the AML co-culture allowed cells to resume differentiation to the proerythroblast stage. These experiments demonstrate that AML imparts a differentiation blockade along the MEP-proerythroblast axis in a cell non-autonomous, reversible fashion. Using cytokine array analysis, we identified elevated IL-6 levels in AML sample-derived CM (n=10, 2841±766.4 pg/ml, 7.80 fold increase, p=0.03) compared to CD34+-derived CM (n=5, 364±36.0 pg/ml). Increased IL-6 was also found in BM aspirates from human AML engrafted splenectomized NSG mice (715±125pg/ml, p=0.001) compared to mice engrafted with normal CD34+ HSPCs (undetectable). Thus, we hypothesized that IL-6 produced by AML blasts acts as a paracrine factor to suppress erythropoiesis. Consistent with this hypothesis, an IL-6 neutralizing antibody reversed the inhibition of BFU-E formation imparted by AML-CM. Furthermore, the addition of recombinant IL-6 to liquid cultures of erythroid differentiation resulted in a 23% reduction in proerythroblasts. Together, our data suggests that (1) overcrowding is not the primary mechanism resulting in BM failure in AML; (2) AML blasts play a previously unrecognized role in imparting a differentiation blockade along the MEP-proerythroblast axis, resulting in progressive anemia; and (3) this differentiation blockade is at least partially attributable to IL-6 secreted from AML blasts as a paracrine factor. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
25. Macrophages from 11β-Hydroxysteroid Dehydrogenase Type 1-Deficient Mice Exhibit an Increased Sensitivity to Lipopolysaccharide Stimulation Due to TGF-β-Mediated Up-Regulation of SHIP1 Expression
- Author
-
Raymond A. Daynes and Tian Y. Zhang
- Subjects
Lipopolysaccharides ,medicine.medical_specialty ,Lipopolysaccharide ,medicine.medical_treatment ,Immunology ,Stimulation ,Sensitivity and Specificity ,Proinflammatory cytokine ,Mice ,chemistry.chemical_compound ,Bone Marrow ,Transforming Growth Factor beta ,Internal medicine ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,medicine ,Animals ,Immunology and Allergy ,Receptor ,Glucocorticoids ,Protein kinase B ,Cells, Cultured ,Mice, Knockout ,B-Lymphocytes ,biology ,Macrophages ,Body Weight ,Inositol Polyphosphate 5-Phosphatases ,Phosphoric Monoester Hydrolases ,Up-Regulation ,Enzyme Activation ,Mice, Inbred C57BL ,Cytokine ,Endocrinology ,chemistry ,Integrin alpha M ,Mink ,Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases ,biology.protein ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Spleen ,Signal Transduction - Abstract
11beta-Hydroxysteroid dehydrogenase type 1 (11betaHSD1) performs end-organ metabolism of glucocorticoids (GCs) by catalyzing the conversion of C(11)-keto-GCs to C(11)-hydroxy-GCs, thereby generating activating ligands for the GC receptor. In this study, we report that 11betaHSD1(-/-) mice are more susceptible to endotoxemia, evidenced by increased weight loss and serum TNF-alpha, IL-6, and IL-12p40 levels following LPS challenge in vivo. Peritoneal and splenic macrophage (splnMphi) from these genetically altered mice overproduce inflammatory cytokines following LPS stimulation in vitro. Inflammatory cytokine overexpression by 11betaHSD1(-/-) splnMphi results from an increased activation of NF-kappaB- and MAPK-signaling cascades and an attenuated PI3K-dependent Akt activation. The expression of SHIP1 is augmented in 11betaHSD1(-/-) Mphi and contributes to inflammatory cytokine production because overexpression of SHIP1 in primary bone marrow Mphi (BMMphi) leads to a similar type of hyperresponsiveness to subsequent LPS stimulation. 11betaHSD1(+/+) and 11betaHSD1(-/-) BMMphi responded to LPS similarly. However, 11betaHSD1(-/-) BMMphi derived in the presence of elevated GC levels up-regulated SHIP1 expression and increased their capacity to produce inflammatory cytokines following their activation with LPS. These observations suggest the hyperresponsiveness of 11betaHSD1(-/-) splnMphi results from myeloid cell differentiation in the presence of moderately elevated GC levels found within 11betaHSD1(-/-) mice. GC-conditioning of BMMphi enhanced SHIP1 expression via up-regulation of bioactive TGF-beta. Consistently, TGF-beta protein expression was increased in unstimulated CD11b(-) cells residing in the BM and spleen of 11betaHSD1(-/-) mice. Our results suggest that modest elevations in plasma GC levels can modify the LPS responsiveness of Mphi by augmenting SHIP1 expression through a TGF-beta-dependent mechanism.
- Published
- 2007
- Full Text
- View/download PDF
26. Analysis on scattering and relationship with granular size in THz spectra
- Author
-
Fang M. Wu, Bing F. Wu, Xiao Y. Zhao, Zhao H. Zhang, Yan Fang, Tian Y. Zhang, and Han Zhang
- Subjects
Materials science ,Optics ,Absorption spectroscopy ,Scattering ,Terahertz radiation ,business.industry ,Mie scattering ,Ranging ,Solid material ,Absorption (electromagnetic radiation) ,business ,Spectral line ,Computational physics - Abstract
Scattering is an important phenomenon and distorts the THz absorption spectrum during analysis of materials. It is essential to understand how scattering from samples changes the THz signals. We presented an approach for estimating the relationship between scattering and granular sizes occurring in THz spectra. The samples were made of same solid material but in different granular sizes ranging from 58um to 150um. The experiment spectra indicated the absorption curves rose up with the granular size increasing, which met the simulation consequence based on Mie scattering. The empirical baseline models were presented in this paper, after removing the baseline, the result indicated that computational accuracy was improved, which can be used in quantitative analysis.
- Published
- 2015
- Full Text
- View/download PDF
27. Peroxisome Proliferator-Activated Receptor α Negatively Regulates T-bet Transcription Through Suppression of p38 Mitogen-Activated Protein Kinase Activation
- Author
-
Raymond A. Daynes, Xiaohong Ding, Dallas C. Jones, and Tian Y. Zhang
- Subjects
CD4-Positive T-Lymphocytes ,Time Factors ,Transcription, Genetic ,T cell ,Immunology ,Down-Regulation ,Receptors, Cytoplasmic and Nuclear ,Mice, Inbred Strains ,Biology ,Mitogen-activated protein kinase kinase ,Ligands ,Lymphocyte Activation ,p38 Mitogen-Activated Protein Kinases ,digestive system ,Interferon-gamma ,Jurkat Cells ,Mice ,medicine ,Animals ,Humans ,Immunology and Allergy ,RNA, Messenger ,IL-2 receptor ,Phosphorylation ,Receptor ,Receptors, Interferon ,Insulin-like growth factor 1 receptor ,Mice, Knockout ,MAP kinase kinase kinase ,Phenylurea Compounds ,ZAP70 ,Up-Regulation ,Cell biology ,Enzyme Activation ,Butyrates ,medicine.anatomical_structure ,Cancer research ,Interleukin-2 ,lipids (amino acids, peptides, and proteins) ,Peroxisome proliferator-activated receptor alpha ,Mitogen-Activated Protein Kinases ,T-Box Domain Proteins ,Signal Transduction ,Transcription Factors - Abstract
Expression of the nuclear hormone receptor peroxisome proliferator-activated receptor α (PPARα) in resting lymphocytes was recently established, although the physiologic role(s) played by this nuclear hormone receptor in these cell types remains unresolved. In this study, we used CD4+ T cells isolated from PPARα−/− and wild-type mice, as well as cell lines that constitutively express PPARα, in experiments designed to evaluate the role of this hormone receptor in the regulation of T cell function. We report that activated CD4+ T cells lacking PPARα produce increased levels of IFN-γ, but significantly lower levels of IL-2 when compared with activated wild-type CD4+ T cells. Furthermore, we demonstrate that PPARα regulates the expression of these cytokines by CD4+ T cells in part, through its ability to negatively regulate the transcription of T-bet. The induction of T-bet expression in CD4+ T cells was determined to be positively influenced by p38 mitogen-activated protein (MAP) kinase activation, and the presence of unliganded PPARα effectively suppressed the phosphorylation of p38 MAP kinase. The activation of PPARα with highly specific ligands relaxed its capacity to suppress p38 MAP kinase phosphorylation and promoted T-bet expression. These results demonstrate a novel DNA-binding independent and agonist-controlled regulatory influence by the nuclear hormone receptor PPARα.
- Published
- 2003
- Full Text
- View/download PDF
28. Glucocorticoid conditioning of myeloid progenitors enhances TLR4 signaling via negative regulation of the phosphatidylinositol 3-kinase-Akt pathway
- Author
-
Raymond A. Daynes and Tian Y. Zhang
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Endogeny ,Stimulation ,Biology ,Proinflammatory cytokine ,Mice ,Phosphatidylinositol 3-Kinases ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Protein kinase B ,Glucocorticoids ,Cells, Cultured ,Myeloid Progenitor Cells ,Gene knockdown ,Macrophages ,Cell Differentiation ,Cell biology ,Toll-Like Receptor 4 ,Haematopoiesis ,Cytokine ,Endocrinology ,Gene Expression Regulation ,Proto-Oncogene Proteins c-akt ,Glucocorticoid ,medicine.drug ,Signal Transduction - Abstract
The immunomodulatory effects of glucocorticoids (GCs) have been described as bimodal, with high levels of GCs exerting immunosuppressive effects and low doses of GCs being immunopermissive. While the mechanisms used by GCs to achieve immunosuppression have been investigated intensely, the molecular mechanisms underlying the permissive effects of GCs remain uncharacterized. Herein, we demonstrate that GC conditioning during the differentiation of myeloid progenitors into macrophages (Mφs) results in their enhanced LPS responsiveness, demonstrated by an overexpression of the inflammatory cytokines TNF-α, IL-6, and IL-12. Inflammatory cytokine overexpression resulted from an increased activation of NF-κB and the MAPK signaling cascade and a reduced activation of the PI3K-Akt pathway following LPS stimulation. GC conditioning during Mφ differentiation induced an increase in the expression of SHIP1, a phosphatase that negatively regulates the PI3K signaling pathway. Small interfering RNA-mediated knockdown of SHIP1 expression increased PI3K-dependent Akt activation and subsequently decreased inflammatory cytokine expression, suggesting GC-mediated up-regulation of SHIP1 expression is responsible for the augmentation in inflammatory cytokine production following LPS stimulation. We also show that splenic Mφs purified from normal mice that were implanted with timed-release GC pellets exhibited an enhanced LPS responsiveness and increased SHIP1 expression, indicating that GCs can regulate SHIP1 expression in vivo. Our results suggest that minor fluctuations in physiological levels of endogenous GCs can program endotoxin-responsive hemopoietic cells during their differentiation by regulating their sensitivity to stimulation.
- Published
- 2007
29. Direct Contact With Bone Marrow Stromal Cells Protects CML Progenitors From Imatinib Through Cytoplasmic Stabilization Of β-Catenin
- Author
-
Anna M. Eiring, David J. Anderson, Tian Y. Zhang, Ira L. Kraft, Kimberly R. Reynolds, Anthony D. Pomicter, Clinton C. Mason, Thomas O'Hare, and Michael W. Deininger
- Subjects
Stromal cell ,Chemistry ,Immunology ,CD34 ,Imatinib ,Cell Biology ,Hematology ,Biochemistry ,medicine.anatomical_structure ,Cell culture ,hemic and lymphatic diseases ,medicine ,Cancer research ,Bone marrow ,Kinase activity ,Progenitor cell ,Tyrosine kinase ,medicine.drug - Abstract
A β-catenin expression signature is associated with primary resistance to tyrosine kinase inhibitors (TKIs) (McWeeney et al. 2010), but its role in TKI resistance is not completely understood. To assess the role of β-catenin in TKI resistance, we used shRNA targeting β-catenin (shβcat) in in vitro models of BCR-ABL1 kinase-independent resistance. To model resistance in the absence of bone marrow (BM)-derived factors, we used TKI-resistant K562R cells that are adapted for growth in the presence of imatinib, as well as primary CD34+ progenitors from CML patients who had failed treatment with two or more TKIs. These cells are cultured in regular medium (RM), proliferate in 1.0-2.5 μM imatinib, and exhibit β-catenin expression that is independent of BCR-ABL1 kinase activity. To model resistance mediated by the BM microenvironment, we cultured TKI-sensitive parental K562 cells and CD34+ progenitors from newly diagnosed CML patients in direct contact (DC) with HS-5 BM stromal cells. HS-5 co-culture increases β-catenin protein levels and clonogenic potential by >3-fold despite continued suppression of BCR-ABL1 kinase activity. All cells lack BCR-ABL1 kinase domain mutations and undergo TKI-mediated kinase inhibition as detected by immunoblot analyses. CML cell lines and primary cells with BCR-ABL1-independent resistance were lentivirally transduced with shβcat or a scrambled control (shSCR), and knockdown was confirmed by immunoblot and/or qRT-PCR. In RM, shβcat reduced the clonogenicity of TKI-sensitive K562 cells and CD34+ cells from newly diagnosed CML patients by 49% and 39%, respectively, compared to shSCR controls. In TKI-resistant K562R cells and CD34+ cells from TKI-resistant CML patients, shβcat reduced clonogenicity by 60% and 50%, respectively, in the presence or absence of imatinib (0-10 μM), suggesting a role for β-catenin in the development or maintenance of TKI resistance. In contrast to cells grown in RM, clonogenicity of cell lines and patient samples cultured in HS-5 DC was unaffected by shβcat compared to imatinib alone. Immunoblot analyses revealed that β-catenin protein levels were fully restored in HS-5 DC, despite the continued presence of shβcat. qRT-PCR revealed that while cells in HS-5 DC have high amounts of β-catenin protein, the mRNA levels remained similar to shβcat-expressing cells cultured in RM, consistent with post-translational stabilization of β-catenin. Importantly, increased β-catenin was not observed when cells were cultured in HS-5 conditioned medium, indicating that stabilization requires DC with the bone marrow stroma. These data are consistent with a role for β-catenin in TKI resistance mediated by DC with the BM microenvironment, similar to a recent report (Zhang et al., 2013). To understand nuclear versus cytoplasmic distribution following HS-5 DC, CD34+ cells from newly diagnosed CML patients were cultured in RM or HS-5 DC for 36 hours and analyzed for β-catenin localization by immunfluorescence. As expected, cells cultured in RM had low levels of β-catenin in the nucleus and cytoplasm that decreased upon treatment with imatinib (2.5 μM). In contrast, cells cultured in HS-5 DC had a marked increase of β-catenin that was unaffected by treatment with imatinib. While detectable in the nucleus, the majority of β-catenin protein was localized in the cytoplasm and at the cell membrane, consistent with its role in cell-cell junctions. Accordingly, in CD34+ cells from newly diagnosed CML patients, an N-cadherin blocking antibody impaired the clonogenic potential of cells cultured in HS-5 DC, with no significant effect on cells grown in RM. Affymetrix Human Gene 1.0 ST arrays revealed high levels of genes encoding the CDH2 and CDH13 cadherins, which may be involved in N-cadherin-mediated β-catenin stabilization, even in the presence of shβcat. Preliminary data also suggests that HS-5 DC reduces luciferase reporter activity from a construct harboring sequential β-catenin binding elements (pGF1-Lef/Tcf-eFGP-luc), further supporting a role for cytoplasmic β-catenin in TKI resistance. These data demonstrate a critical role for β-catenin in BCR-ABL1 kinase-independent TKI resistance, and suggest new strategies for targeting TKI resistance in the absence of BCR-ABL1 kinase domain mutations. Disclosures: Deininger: Bristol-Myers Squibb: Advisory Boards Other, Consultancy, Research Funding; Ariad Pharmaceuticals: Advisory Boards, Advisory Boards Other, Consultancy; Novartis: Advisory Boards, Advisory Boards Other, Consultancy, Research Funding; Celgene: Research Funding; Gilead Sciences: Research Funding.
- Published
- 2013
- Full Text
- View/download PDF
30. BP5-087, a Novel STAT3 Inhibitor, Combines With BCR-ABL1 Inhibition To Overcome Kinase-Independent Resistance In Chronic Myeloid Leukemia
- Author
-
Anna M. Eiring, Ira L. Kraft, Brent D.G. Page, Tian Y. Zhang, Jamshid S. Khorashad, Nadeem A. Vellore, Kimberly R. Reynolds, Anthony D. Pomicter, Anna V. Senina, Matthew S. Zabriskie, Shazia Ahmad, Clinton C. Mason, Richard Moriggl, Riccardo Baron, Thomas O'Hare, Patrick T. Gunning, and Michael W. Deininger
- Subjects
Kinase ,Chemistry ,medicine.drug_class ,Cell growth ,Immunology ,Myeloid leukemia ,Imatinib ,Cell Biology ,Hematology ,CD38 ,Biochemistry ,Tyrosine-kinase inhibitor ,Imatinib mesylate ,hemic and lymphatic diseases ,medicine ,Cancer research ,Stem cell ,medicine.drug - Abstract
Mutations in the BCR-ABL1 kinase domain are a well-established mechanism of tyrosine kinase inhibitor (TKI) resistance, but fail to explain many cases of clinical TKI failure. In the remaining patients, resistance occurs via activation of alternative signaling pathways that maintain survival despite BCR-ABL1 inhibition (BCR-ABL1-independent resistance). STAT3 mediates TKI resistance in chronic myeloid leukemia (CML) cells cultured in the presence of bone marrow-derived factors (Bewry et al., 2008; Traer et al., 2012; Nair et al., 2012), and also plays a critical role in survival of CML cells with BCR-ABL1-independent resistance (Eiring et al. #31, ASH 2012). While targeting transcription factors is notoriously difficult, our combination of synthetic chemistry, in vitro reporter assays, and computational modeling has led to a low micromolar mechanism-based STAT3 inhibitor, which, in combination with TKIs, shows promise as a treatment for CML patients with BCR-ABL1-independent resistance. The original compound of the series, SF1-066 (10 µM; Fletcher et al., 2009), combines with TKIs to reduce survival of CML CD34+ cells exhibiting BCR-ABL1-independent resistance (Eiring et al. #31, ASH 2012). To improve the potency and selectivity of SF1-066, we synthesized successive STAT3 inhibitor libraries and ranked candidates by structure-activity relationship using a luciferase-based reporter screen (Kraft et al. #2445, ASH 2012). This reporter assay quantifies STAT3 transcriptional activity in TKI-resistant AR230R cells, which grow in the continuous presence of imatinib (1.0 µM), lack BCR-ABL1 kinase domain mutations, and exhibit high levels of pSTAT3Y705, thereby enabling convenient, high-throughput screening for potency and selectivity in the context of endogenous STAT3 activation. Among three sequential STAT3 inhibitor libraries, BP5-087 emerged as the new lead compound. Fluorescence polarization assays verified that BP5-087 was 5-fold more effective than SF1-066 in outcompeting an SH2 peptide probe, and computational simulations predicted better overall binding of BP5-087 (-9.6 kcal/mol) versus SF1-066 (-7.6 kcal/mol) to the STAT3 SH2 interface. In AR230R cell growth assays, BP5-087 was effective at a 5-fold lower dose compared to SF1-066, with minimal effects on TKI-sensitive parental controls. Therefore, we tested BP5-087 in the context of primary TKI resistance. BP5-087 (1 µM) in combination with imatinib (2.5 µM) reduced colony formation and increased apoptosis of CD34+ cells from CML patients with BCR-ABL1-independent resistance. These cells have no BCR-ABL1 kinase domain mutations and undergo BCR-ABL1 kinase inhibition as detected by immunoblot analyses. In contrast, BP5-087 had no effect on CD34+ cells from newly diagnosed CML patients or normal individuals. Immunofluorescence demonstrated that dual treatment of TKI-resistant CML CD34+ cells resulted in reduced levels of nuclear pSTAT3Y705, consistent with an inhibitor of STAT3 dimerization. In more primitive CML stem cells, long term culture-initiating cell (LTC-IC) assays revealed that neither inhibitor alone had any effect on colony formation of primitive LTC-IC progenitors, whereas imatinib (2.5 µM) in combination with BP5-087 (1.0 µM) reduced LTC-IC colony formation by 66%. Consistent with this observation, immunofluorescence showed high levels of pSTAT3Y705 in primitive TKI-resistant CD34+CD38- cells when cultured in the presence but not absence of TKIs. To test the feasibility of BP5-087 for in vivo use, we treated mice orally with BP5-087 (25 mg/kg/day) for 4 weeks and observed no changes in body weight, peripheral blood cellularity, or bone marrow colony forming ability. Mass spectrometry confirmed that BP5-087 is orally bioavailable. In summary, BP5-087 is a systematically-derived, direct inhibitor of STAT3 that, in combination with TKIs, reduces survival of CML cells with BCR-ABL1-independent resistance. Further rounds of structure-activity optimization may reveal an inhibitor with a clinically-relevant effective concentration. Disclosures: Deininger: Bristol Myers Squibb: Advisory Boards Other, Consultancy, Research Funding; Ariad Pharmaceuticals: Advisory Boards, Advisory Boards Other, Consultancy; Novartis: Advisory Boards, Advisory Boards Other, Consultancy, Research Funding; Celgene: Research Funding; Gilead Sciences: Research Funding.
- Published
- 2013
- Full Text
- View/download PDF
31. Suppression of CML Progenitor but Not Stem Cells Requires Simultaneous Inhibition of KIT and BCR-ABL1
- Author
-
Anna M. Eiring, Amie S. Corbin, Tian Y. Zhang, Michael W. Deininger, Brian J. Druker, Zhimin Gu, and Thomas O'Hare
- Subjects
Immunology ,CD34 ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Imatinib mesylate ,Cell culture ,hemic and lymphatic diseases ,Kinase activity ,Stem cell ,Progenitor cell ,Tyrosine kinase ,Interleukin 3 - Abstract
Abstract 2778 In chronic myeloid leukemia (CML), imatinib and other tyrosine kinase inhibitors (TKIs) inhibit BCR-ABL1 tyrosine kinase activity but also target additional kinases including KIT. The role of KIT inhibition in the therapeutic efficacy of TKIs is controversial. We used TKIs with selective activity against ABL (PPY-A) or KIT (BAW667) and genetic tools to assess the role of KIT signaling for growth of CML cell lines and primary CML progenitor and stem cells. In Mo7eBCR-ABL1 or newly diagnosed CML CD34+ progenitor cells, immunoblotting confirmed that PPY-A (1 μM) suppresses BCR-ABL1 phosphorylation but not KIT tyrosine phosphorylation. In contrast, treatment of cells with a KIT-blocking antibody (K44.2, 200ng/mL), shRNA targeting KIT (shKIT), or the KIT selective inhibitor BAW667 (1 μM), suppressed KIT activity without affecting BCR-ABL1 kinase activity. Therefore, these systems are suitable to isolate the role of BCR-ABL1 vs. KIT inhibition. Treatment of Mo7eBCR-ABL1 cells with PPY-A resulted in suppression of growth by 91.7% (p To assess BCR-ABL1 vs. KIT inhibition in primary cells, CD34+ cells from newly diagnosed CML patients (n=4) and normal controls (n=3) were cultured in semisolid medium supplied with IL-3 and GM-CSF (no SCF), in the presence of 1 μM PPY-A combined with shKIT or 1 μM BAW667. KIT inhibition by shKIT or 1 μM BAW667 reduced CFU-GM formation by 40% compared to controls (p To assess the role of KIT vs. BCR-ABL1 inhibition on primitive CML cells we performed long-term culture-initiating cell (LTC-IC) assays on M2–10B4 murine stromal cells, using Lin− cells from newly diagnosed patients (n=3). Cultures were performed with K44.2, PPY-A, K44.2 plus PPY-A or 2 mM imatinib, with colonies plated at 1, 3, and 6 weeks. At 1 week colonies were reduced by 30% with K44.2 and 70% with PPY-A, but by 90% with the PPY-A / K44.2 combination or with imatinib. In contrast, at 6 weeks colony formation was unaffected by K44.2 but reduced by >95% with PPY-A, the PPY-A / K44.2 combination or imatinib. Week 3 colony growth was intermediate. Consistent with the LTC-IC assay, KIT inhibition with BAW667 enhanced PPY-A suppression of colony formation in Lin−CD34+CD38+ progenitor cells from newly diagnosed patients (n=3) by 18.7% (p Our findings suggest KIT inhibition is much more critical for suppression of mature progenitors compared to primitive CML cells. Since AKT is active in CML progenitors but suppressed by TGFβ in stem cells (Nature, 2010;463(7281):676; JCI, 2011;121(1):396), we speculate that upon BCR-ABL1 inhibition CML progenitors but not stem cells switch to an SCF-dependent mode of AKT activation, which renders these cells uniquely sensitive to dual inhibition of BCR-ABL1 and KIT signaling. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
- Full Text
- View/download PDF
32. Hepcidin mediates transcriptional changes that modulate acute cytokine-induced inflammatory responses in mice
- Author
-
Nyall London, Dean Y. Li, Jerry Kaplan, Diane M. Ward, Curry L. Koening, Ryan W. Branch, James P. Kushner, Ivana De Domenico, Eric Lo, Raymond A. Daynes, and Tian Y. Zhang
- Subjects
inorganic chemicals ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,medicine.medical_treatment ,Ferroportin ,digestive system ,Hepcidin ,hemic and lymphatic diseases ,Internal medicine ,Clinical investigation ,Medicine ,STAT3 ,Protein kinase A ,Transcription factor ,biology ,business.industry ,nutritional and metabolic diseases ,General Medicine ,STAT3 Transcription Factor ,Cell biology ,Endocrinology ,Cytokine ,Immunology ,biology.protein ,Tumor necrosis factor alpha ,Corrigendum ,business - Abstract
Hepcidin is a peptide hormone that regulates iron homeostasis and acts as an antimicrobial peptide. It is expressed and secreted by a variety of cell types in response to iron loading and inflammation. Hepcidin mediates iron homeostasis by binding to the iron exporter ferroportin, inducing its internalization and degradation via activation of the protein kinase Jak2 and the subsequent phosphorylation of ferroportin. Here we have shown that hepcidin-activated Jak2 also phosphorylates the transcription factor Stat3, resulting in a transcriptional response. Hepcidin treatment of ferroportin-expressing mouse macrophages showed changes in mRNA expression levels of a wide variety of genes. The changes in transcript levels for half of these genes were a direct effect of hepcidin, as shown by cycloheximide insensitivity, and dependent on the presence of Stat3. Hepcidin-mediated transcriptional changes modulated LPS-induced transcription in both cultured macrophages and in vivo mouse models, as demonstrated by suppression of IL-6 and TNF-α transcript and secreted protein. Hepcidin-mediated transcription in mice also suppressed toxicity and morbidity due to single doses of LPS, poly(I:C), and turpentine, which is used to model chronic inflammatory disease. Most notably, we demonstrated that hepcidin pretreatment protected mice from a lethal dose of LPS and that hepcidin-knockout mice could be rescued from LPS toxicity by injection of hepcidin. The results of our study suggest a new function for hepcidin in modulating acute inflammatory responses.
- Published
- 2012
- Full Text
- View/download PDF
33. Cross-axis synchronous flow-through coil planet centrifuge for large-scale preparative counter-current chromatography
- Author
-
Yoichiro Ito and Tian Y. Zhang
- Subjects
Centrifuge ,Chromatography ,Countercurrent chromatography ,Electromagnetic coil ,Chemistry ,Stationary phase ,Planet ,Organic Chemistry ,General Medicine ,Biochemistry ,Analytical Chemistry ,Radial plane - Abstract
A countercurrent chromatography apparatus and method where the column rotates about an axis spaced apart from, parallel to, and in the same radial plane as a radius extending from the central axis of revolution. The apparatus generates a unique force field which enables excellent separation.
- Published
- 1988
- Full Text
- View/download PDF
34. KIT Signaling Governs Differential Sensitivity of Mature and Primitive CML Progenitors to Tyrosine Kinase Inhibitors.
- Author
-
Corbin, Amie S., O'Hare, Thomas, Zhimin Gu, Kraft, Ira L., Eiring, Anna M., Khorashad, Jamshid S., Pomicter, Anthony D., Tian Y. Zhang, Eide, Christopher A., Manley, Paul W., Cortes, Jorge E., Druker, Brian J., and Deininger, Michael W.
- Subjects
- *
PROTEIN-tyrosine kinase inhibitors , *TREATMENT of chronic myeloid leukemia , *PROGENITOR cells , *STEM cell treatment , *DRUG therapy , *IMATINIB - Abstract
Imatinib and other BCR-ABL1 inhibitors are effective therapies for chronic myelogenous leukemia (CML), but these inhibitors target additional kinases including KIT, raising the question of whether off-target effects contribute to clinical efficacy. On the basis of its involvement in CML pathogenesis, we hypothesized that KIT may govern responses of CML cells to imatinib. To test this, we assessed the growth of primary CML progenitor cells under conditions of sole BCR-ABL1, sole KIT, and dual BCR-ABL1/KIT inhibition. Sole BCR-ABL1 inhibition suppressed mature CML progenitor cells, but these effects were largely abolished by stem cell factor (SCF) and maximal suppression required dual BCR-ABL1/KIT inhibition. In contrast, KIT inhibition did not add to the effects of BCR-ABL1 inhibition in primitive progenitors, represented by CD34+ 38- cells. Long-term culture-initiating cell assays on murine stroma revealed profound depletion of primitive CML cells by sole BCR-ABL1 inhibition despite the presence of SCF, suggesting that primitive CML cells are unable to use SCF as a survival factor upon BCR-ABL1 inhibition. In CD34+38+ cells, SCF strongly induced pAKTS473 in a phosphoinositide kinase (PI3K)--dependent manner, which was further enhanced by inhibition of BCR-ABL1 and associated with increased colony survival. In contrast, pAKTS473+ levels remained low in CD34-38- cells cultured under the same conditions. Consistent with reduced response to SCF, KIT surface expression was significantly lower on CD34+ 38- compared with CD34+38- CML cells, suggesting a possible mechanism for the differential effects of SCF on mature and primitive CML progenitor cells. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.