Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nikolai A. Podoltsev is active.

Publication


Featured researches published by Nikolai A. Podoltsev.


Nature Genetics | 2014

Mutation of NLRC4 causes a syndrome of enterocolitis and autoinflammation

Neil Romberg; Khatoun Al Moussawi; Carol Nelson-Williams; Amy L. Stiegler; Erin Loring; Murim Choi; John D. Overton; Eric Meffre; Mustafa K. Khokha; Anita J Huttner; Brian L. West; Nikolai A. Podoltsev; Titus J. Boggon; Barbara I. Kazmierczak; Richard P. Lifton

Upon detection of pathogen-associated molecular patterns, innate immune receptors initiate inflammatory responses. These receptors include cytoplasmic NOD-like receptors (NLRs) whose stimulation recruits and proteolytically activates caspase-1 within the inflammasome, a multiprotein complex. Caspase-1 mediates the production of interleukin-1 family cytokines (IL1FCs), leading to fever and inflammatory cell death (pyroptosis). Mutations that constitutively activate these pathways underlie several autoinflammatory diseases with diverse clinical features. We describe a family with a previously unreported syndrome featuring neonatal-onset enterocolitis, periodic fever, and fatal or near-fatal episodes of autoinflammation. We show that the disease is caused by a de novo gain-of-function mutation in NLRC4 encoding a p.Val341Ala substitution in the HD1 domain of the protein that cosegregates with disease. Mutant NLRC4 causes constitutive IL1FC production and macrophage cell death. Infected macrophages from affected individuals are polarized toward pyroptosis and exhibit abnormal staining for inflammasome components. These findings identify and describe the cause of a life-threatening but treatable autoinflammatory disease that underscores the divergent roles of the NLRC4 inflammasome.


Cancer Investigation | 2009

Phase II Clinical Trial of Paclitaxel Loaded Polymeric Micelle in Patients with Advanced Pancreatic Cancer

Muhammad Wasif Saif; Nikolai A. Podoltsev; Mark S. Rubin; Jose A. Figueroa; Min-Young Lee; Jin-Won Kwon; Elin Rowen; Jaewon Yu; Robert O. Kerr

ABSTRACT Purpose: To determine in patients, with locally advanced or metastatic pancreatic cancer (APC), efficacy and safety of treatment with intravenous paclitaxel loaded polymeric micelle (GPM). Patients and methods: This was a multicenter, open-label Phase II study. Patients with APC, ECOG performance status ≤ 2, no prior chemotherapy and adequate organ function were treated with 3-hour GPM infusions every 3 weeks. Initial patients were treated with 435 mg/m2 (n = 11). The dose was reduced for subsequent patients to 350 or 300 mg/m2 (n = 45). Primary endpoint was time to tumor progression (TTP). Results: 56 patients were enrolled. Median TTP for patients treated with 300 or 350 mg/m2 doses was 3.2 months (95% CI, 2.6–4.2). Median progression free survival (PFS) was 2.8 months (95% CI, 1.4–4.0). Median overall survival (OS) was 6.5 months (95% CI, 5.1–7.9). Among patients treated with above doses of GPM, there was 1 complete remission (CR) and 2 partial remissions (PR) with an overall response rate (ORR) of 6.7%. Disease control rate (CR + PR + stable disease) was 60.0%. Most common grade 3 toxicities were: neutropenia (40.0%), fatigue (17.8%), infection, dehydration, neuropathy (each 13.3%), and abdominal pain (11.1%). Conclusions: Treatment of APC with GPM at a dose of 300 mg/m2 q 3 weeks was well tolerated and common toxicities were qualitatively similar to Cremophor-based paclitaxel. GPM monotherapy resulted in OS and other efficacy parameters preferable to that seen historically with gemcitabine. Future studies of GPM in combination with other agents for treatment of APC are warranted.


American Journal of Clinical Pathology | 2014

Marrow assessment for hemophagocytic lymphohistiocytosis demonstrates poor correlation with disease probability.

Caleb Ho; Xiaopan Yao; Ligeng Tian; Fangyong Li; Nikolai A. Podoltsev; Mina L. Xu

OBJECTIVES To evaluate the amount of hemophagocytosis in 64 marrow core biopsy specimens and aspirates from 58 patients with clinical suspicion for secondary hemophagocytic lymphohistiocytosis (HLH) or reported findings of hemophagocytosis. METHODS A review of medical records assigned patients to a low-risk (45 patients) or high-risk (13 patients) HLH group, and association with histologic findings was examined using the Fisher exact test. RESULTS The amount of hemophagocytosis in aspirate or the core biopsy specimen did not correlate with disease probability (P = .17 and P = .63, respectively). Of the clinical/laboratory criteria assessed, the most significant correlations with HLH were highly elevated ferritin (P = .01), cytopenias (P = .02), and fever (P = .009). CONCLUSIONS Our findings indicated that marrow histologic findings alone do not reliably predict the probability of HLH, and an isolated finding of hemophagocytosis, even when present in a high amount, lacks specificity for HLH.


Clinical Colorectal Cancer | 2009

Oxaliplatin-Induced Immune Thrombocytopenia: Another Cumulative Dose-Dependent Side Effect?

Edward James; Nikolai A. Podoltsev; Elahe Salehi; Brian R. Curtis; Muhammad Wasif Saif

Oxaliplatin is a third-generation platinum compound that is commonly used for the treatment of colorectal cancer (CRC) both in the adjuvant and metastatic disease settings. Oxaliplatin-based chemotherapy is presently limited by cumulative dose-dependent neurotoxicity. We had previously reported on 2 patients who developed oxaliplatin-induced immune thrombocytopenia (OITP) during their tenth and seventeenth FOLFOX (5-fluorouracil/leucovorin/ oxaliplatin) cycles. Herein, we report on a third patient who developed severe thrombocytopenia after 28 cycles of modified FOLFOX6 (mFOLFOX) chemotherapy. A 60-year-old white woman with metastatic CRC, who had partial sigmoidectomy and colostomy, presented with bleeding from stoma site, on cycle 28, day 1 of mFOLFOX6, 7.5 hours after completion of the oxaliplatin infusion. Laboratory data revealed marked thrombocytopenia with a nadir platelet count of zero. Due to concern for OITP, the patients serum was sent to BloodCenter of Wisconsin, Inc. (Diagnostic Laboratories; Milwaukee, WI). Serologic testing for oxaliplatin-dependent platelet antibodies was performed using flow cytometry in the presence of various concentrations of oxaliplatin. Laboratory tests for autoimmune hemolysis and disseminated intravascular coagulation (DIC) were negative. Oxaliplatin-dependent platelet reactive antibodies were detected in the patients serum, confirming the diagnosis of OITP. The diagnosis of OITP should be entertained in patients receiving oxaliplatin for prolonged periods of time even though there are other more common causes of thrombocytopenia in these patients, including chemotherapy-induced myelosuppression, bone marrow involvement with tumor, and DIC.


Leukemia Research | 2012

Proposed criteria for response assessment in patients treated in clinical trials for myeloproliferative neoplasms in blast phase (MPN-BP): Formal recommendations from the post-myeloproliferative neoplasm acute myeloid leukemia consortium

John Mascarenhas; Mark L. Heaney; Vesna Najfeld; Elizabeth O. Hexner; Omar Abdel-Wahab; Raajit Rampal; Farhad Ravandi; Bruce Petersen; Gail J. Roboz; Eric J. Feldman; Nikolai A. Podoltsev; Dan Douer; Ross L. Levine; Martin S. Tallman; Ronald Hoffman

Leukemic transformation (LT) of a myeloproliferative neoplasm (MPN) is associated with a dismal prognosis and no medical therapies have shown a survival improvement in patients with MPN in blast phase (MPN-BP). Effective therapies for the treatment of MPN-BP are a serious unmet need. Consensus response criteria do not exist for the treatment of patients with MPN-BP and this is necessary for the uniformed reporting of treatment response in clinical trials. We have identified relevant MPN and MPN-BP features in order to define treatment response categories that reflect hematological, clinical, pathological, cytogenetic and molecular changes after therapeutic intervention. We plan to validate these proposed response criteria within multi-centered clinical trials.


British Journal of Haematology | 2016

Comparative clinical effectiveness of azacitidine versus decitabine in older patients with myelodysplastic syndromes.

Amer M. Zeidan; Amy J. Davidoff; Jessica B. Long; Xin Hu; Rong Wang; Xiaomei Ma; Cary P. Gross; Gregory A. Abel; Scott F. Huntington; Nikolai A. Podoltsev; Uno Hajime; Thomas Prebet; Steven D. Gore

The hypomethylating agents (HMAs) azacitidine and decitabine are both approved for treatment of myelodysplastic syndromes (MDS) in the USA. In Europe, decitabine is not approved due to lack of survival advantage in randomized trials. The two drugs have not been compared in clinical trials. We identified patients diagnosed with MDS between 2004 and 2011 from the Surveillance, Epidemiology, and End Results (SEER)‐Medicare linked database in the USA who received ≥ 10 doses of either HMA. We estimated survival from HMA initiation with Kaplan–Meier methods and used multivariate Cox proportional hazards models to adjust for covariates. Analyses controlled for histological subtype and we conducted a subset analysis limited to patients with refractory anaemia with excess blasts (RAEB). In 2025 HMA‐treated patients, median survival was 15 months with no difference in survival based on the HMA received in adjusted analysis (decitabine versus azacitidine, hazard ratio = 1·06, 95% confidence interval: 0·94–1·19, P = 0·37). For RAEB patients (n = 523), median survival was 12 months, with no significant difference based on HMA received. No significant survival difference was found between azacitidine and decitabine in patients with MDS, including RAEB. Importantly, population‐based survival of azacitidine‐treated RAEB patients was substantially shorter than in the AZA‐001 clinical trial (11 versus 24·5 months).


Lancet Oncology | 2017

Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: phase 2 results from a multicentre, randomised, phase 1/2 trial

Hagop M. Kantarjian; Gail J. Roboz; Patricia Kropf; Karen Yee; Casey O'Connell; Raoul Tibes; Katherine Walsh; Nikolai A. Podoltsev; Elizabeth A. Griffiths; Elias Jabbour; Guillermo Garcia-Manero; David A. Rizzieri; Wendy Stock; Michael R. Savona; Todd L. Rosenblat; Jesus G. Berdeja; Farhad Ravandi; Edwin P. Rock; Yong Hao; Mohammad Azab; Jean-Pierre Issa

BACKGROUND The hypomethylating drugs azacitidine and decitabine have shown efficacy in myelodysplastic syndromes and acute myeloid leukaemia, but complete tumour responses are infrequent and of short duration, possibly because of the short half-lives and suboptimal bone marrow exposure of the drugs. Guadecitabine, a next-generation hypomethylating drug, has a longer half-life and exposure than its active metabolite decitabine. A phase 1 study established 60 mg/m2 guadecitabine for 5 days as an effective treatment schedule. In this phase 2 study, we aimed to assess the safety and activity of two doses and schedules of guadecitabine in older (≥65 years) patients with treatment-naive acute myeloid leukaemia who were not candidates for intensive chemotherapy. METHODS We did a multicentre, randomised, open-label, phase 1/2 study of guadecitabine in cohorts of patients with treatment-naive acute myeloid leukaemia, relapsed or refractory acute myeloid leukaemia, and myelodysplastic syndromes; here we report the phase 2 results from the cohort of treatment-naive patients with acute myeloid leukaemia. We included patients aged at least 65 years from 14 US medical centres (hospitals and specialist cancer clinics) who were not candidates for intensive chemotherapy and randomly assigned them (1:1) using a computer algorithm (for dynamic randomisation) to guadecitabine 60 or 90 mg/m2 on days 1-5 (5-day schedule) of a 28-day treatment cycle. Treatment allocation was not masked. We also assigned additional patients to guadecitabine 60 mg/m2 in a 10-day schedule in a 28-day treatment cycle after a protocol amendment. The primary endpoint was composite complete response (complete response, complete response with incomplete platelet recovery, or complete response with incomplete neutrophil recovery regardless of platelets). Response was assessed in all patients (as-treated) who received at least one dose of guadecitabine. We present the final analysis, although at the time of the database lock, 15 patients were still in follow-up for overall survival. This study is registered with ClinicalTrials.gov, number NCT01261312. FINDINGS Between Aug 24, 2012, and Sept 15, 2014, 107 patients were enrolled: 54 on the 5-day schedule (26 randomly assigned to 60 mg/m2 and 28 to 90 mg/m2) and 53 were assigned to the 10-day schedule. Median age was 77 years (range 62-92), and median follow-up was 953 days (IQR 721-1040). All treated patients were assessable for a response. The number of patients who achieved a composite complete response did not differ between dose groups or schedules (13 [54%, 95% CI 32·8-74·4] with 60 mg/m2 on the 5-day schedule; 16 [59%; 38·8-77·6] with 90 mg/m2 on the 5-day schedule; and 26 [50%, 35·8-64·2] with 60 mg/m2 on the 10-day schedule). The most frequent grade 3 or worse adverse events, regardless of relationship to treatment, were febrile neutropenia (31 [61%] of 51 patients on the 5-day schedule vs 36 [69%] of 52 patients on the 10-day schedule), thrombocytopenia (25 [49%] vs 22 [42%]), neutropenia (20 [39%] vs 18 [35%]), pneumonia (15 [29%] vs 19 [37%]), anaemia (15 [29%] vs 12 [23%]), and sepsis (eight [16%] vs 14 [27%]). The most common serious adverse events, regardless of relationship to treatment, for the 5-day and 10-day schedules, respectively, were febrile neutropenia (27 [53%] vs 25 [48%]), pneumonia (14 [27%] vs 16 [31%]), and sepsis (eight [16%] vs 14 [27%]). 23 (22%) patients died because of adverse events (mainly from sepsis, eight [8%]; and pneumonia, five [5%]); four deaths were from adverse events deemed treatment-related (pneumonia, two [2%]; multiorgan failure, one [1%]; and sepsis, one [1%], all in the 10-day cohort). INTERPRETATION More than half of older treatment-naive patients with acute myeloid leukaemia achieved a composite complete response with guadecitabine at all drug doses and schedules investigated, with tolerable toxicity. The recommended guadecitabine regimen for this population is 60 mg/m2 in a 5-day schedule. A phase 3 study in this patient population is ongoing (NCT02348489) to assess guadecitabine 60 mg/m2 in a 5-day schedule versus standard of care. FUNDING Astex Pharmaceuticals and Stand Up To Cancer.


Blood Reviews | 2016

The evolving field of prognostication and risk stratification in MDS: Recent developments and future directions

Eun-Ju Lee; Nikolai A. Podoltsev; Steven D. Gore; Amer M. Zeidan

The clinical course of patients with myelodysplastic syndromes (MDS) is characterized by wide variability reflecting the underlying genetic and biological heterogeneity of the disease. Accurate prediction of outcomes for individual patients is an integral part of the evidence-based risk/benefit calculations that are necessary for tailoring the aggressiveness of therapeutic interventions. While several prognostication tools have been developed and validated for risk stratification, each of these systems has limitations. The recent progress in genomic sequencing techniques has led to discoveries of recurrent molecular mutations in MDS patients with independent impact on relevant clinical outcomes. Reliable assays of these mutations have already entered the clinic and efforts are currently ongoing to formally incorporate mutational analysis into the existing clinicopathologic risk stratification tools. Additionally, mutational analysis holds promise for going beyond prognostication to therapeutic selection and individualized treatment-specific prediction of outcomes; abilities that would revolutionize MDS patient care. Despite these exciting developments, the best way of incorporating molecular testing for use in prognostication and prediction of outcomes in clinical practice remains undefined and further research is warranted.


Clinical Colorectal Cancer | 2011

Mitomycin-C-Induced Thrombotic Thrombocytopenic Purpura/Hemolytic Uremic Syndrome: Cumulative Toxicity of an Old Drug in a New Era

Roland El-Ghazal; Nikolai A. Podoltsev; Peter W. Marks; Edward Chu; Muhammad Wasif Saif

Mitomycin-C (MMC) is a first-line therapy for anal squamous cell carcinoma (ASCC), and it continues to be used for several other indications. Thrombotic thrombocytopenic purpura (TTP) and hemolytic uremic syndrome (HUS) are serious complications of MMC therapy. Herein, we describe a 73-year-old woman with recurrent ASCC treated with MMC who developed chronic microangiopathic hemolytic anemia (MAHA) and thrombocytopenia after receiving a cumulative dose of 50 mg/m(2). These hematologic adverse events persisted for 9 months and resolved after discontinuation of chemotherapy followed by multiple courses of oral corticosteroids. This report describes an atypically chronic and mild form of secondary TTP/HUS and discusses the possible role of corticosteroids in its management.


Expert Review of Anticancer Therapy | 2015

The clinical use of DNA methyltransferase inhibitors in myelodysplastic syndromes

Abou Zahr A; Saad Aldin E; Lisa Barbarotta; Nikolai A. Podoltsev; Amer M. Zeidan

Myelodysplastic syndromes (MDS) include a heterogeneous group of acquired hematopoietic malignancies characterized by ineffective hematopoiesis, peripheral cytopenias, and a varying propensity for progression to acute myeloid leukemia. Patients with higher risk MDS have dismal outcomes and treatment options are very limited. Aside from allogeneic hematopoietic cell transplantation, the only potentially curative treatment, DNA methyltransferase inhibitors (DNMTIs) are the only intervention that prolongs overall survival in patients with higher risk MDS. The clinical use of DNMTIs in MDS was one of the earliest successful attempts at epigenetic reprogramming of cancer. In this review, we discuss the clinical use of DNMTIs in MDS highlighting the current challenges and controversies and future directions of research needed to explore the full therapeutic potential of these agents.

Collaboration


Dive into the Nikolai A. Podoltsev's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rami S. Komrokji

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge