13 results on '"Rudek M"'
Search Results
2. Off-label, but on target:the evidence needed to implement alternative dosing regimens of anticancer drugs
- Author
-
Overbeek, J. K., Heine, R. ter, Verheul, H. M.W., Chatelut, E., Rudek, M. A., Gurney, H., Plummer, R., Gilbert, D. C., Buclin, T., Burger, D. M., Bloemendal, H. J., van Erp, N. P., Overbeek, J. K., Heine, R. ter, Verheul, H. M.W., Chatelut, E., Rudek, M. A., Gurney, H., Plummer, R., Gilbert, D. C., Buclin, T., Burger, D. M., Bloemendal, H. J., and van Erp, N. P.
- Published
- 2023
3. 1833P Concurrent high-dose IV Vitamin C (IVC) and docetaxel for metastatic castrate-resistant prostate cancer (mCRPC): A randomized, placebo-controlled, double-blind phase II trial
- Author
-
Paller, C., Zahurek, M., Mandl, A., Rudek, M., Heath, E., Marshall, C.H., Markowski, M.C., Lalji, A., Metri, N., Hoimes, C.J., Taksey, J., Durham, J., Kelly, W., Antonarakis, E.S., Eisenberger, M., Carducci, M.A., Denmeade, S., and Levine, M.
- Published
- 2023
- Full Text
- View/download PDF
4. Assay for the quantification of abemaciclib, its metabolites, and olaparib in human plasma by liquid chromatography-tandem mass spectrometry.
- Author
-
Hill KL, Abbott NL, Na JY, Rudek M, Moore K, Lee EQ, and Phelps MA
- Subjects
- Humans, Reproducibility of Results, Chromatography, Liquid methods, Limit of Detection, Chromatography, High Pressure Liquid methods, Spectrometry, Mass, Electrospray Ionization methods, Tandem Mass Spectrometry methods, Piperazines blood, Piperazines pharmacokinetics, Aminopyridines blood, Aminopyridines pharmacokinetics, Phthalazines blood, Phthalazines pharmacokinetics, Benzimidazoles blood, Benzimidazoles pharmacokinetics
- Abstract
An isotope-dilution bioanalytical assay for abemaciclib and its metabolites in combination with olaparib was developed and validated in human plasma K2 EDTA. For the quantitative assay, human plasma samples (or human plasma QC samples) were spiked with internal standard solution before a simple protein precipitation with methanol. The extract was injected onto a liquid chromatography-tandem mass spectrometry (LC-MS/MS) instrument where it was chromatographically separated by a polar end-capped reversed phase column and guard using gradient elution with water and methanol both modified with 0.2 % formic acid (v/v) as the mobile phases. The analytes and internal standards were measured by heated electrospray ionization (HESI) in positive polarity using selected reaction monitoring (SRM) on a triple quadrupole mass spectrometer. The assay was validated for linear ranges as follows: 0.4 - 1000 nM abemaciclib, 0.35 - 1000 nM M2 and M18, 0.5 - 1000 nM M20, and 0.75 - 1000 nM olaparib. The inter-day or between day precision for the quality controls (n = 18) was < 13 % and the accuracy was ± 12 %, for all analytes, including the lower limit of quantification (LLOQ). The intra-day or within day precision for the quality controls (n = 6) was ≤ 11 % and the accuracy was ± 12 % for low, mid, and high and < 19 % at LLOQ. The recovery in human plasma was determined to be between 92 % and 102 % for all analytes spanning the linear range. The validated, bioanalytical quantitative assay was designed to measure abemaciclib, its metabolites, and olaparib for pharmacokinetic evaluation of patients in clinical trials for breast, brain, and ovarian cancers., Competing Interests: Declaration of Competing Interest Kathleen Moore: Consultation or advisory fees from Astra Zeneca, Aravive, Aadi, Blueprint, Clovis, Caris, Duality, Eisai, GSK, Genentech/Roche, Immunogen, Mersana, Merck, Myriad, Novartis, Janssen, Regeneron, VBL Theraeputics, Verastem, and Zentalis. Other activities that could influence the work include GOG Foundation BOD, and ASCO BOD., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
5. A multicenter, phase 1, Adult Brain Tumor Consortium trial of oral terameprocol for patients with recurrent high-grade glioma (GATOR).
- Author
-
Ahluwalia MS, Ozair A, Rudek M, Ye X, Holdhoff M, Lieberman FS, Piotrowski AF, Nabors B, Desai A, Lesser G, Huang RC, Glenn S, Khosla AA, Peereboom DM, Wen PY, and Grossman SA
- Subjects
- Humans, Male, Middle Aged, Adult, Female, Aged, Administration, Oral, Aged, 80 and over, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Neoplasm Grading, Maximum Tolerated Dose, Glioma drug therapy, Glioma pathology, Brain Neoplasms drug therapy, Brain Neoplasms pathology
- Abstract
Recurrent high-grade gliomas (rHGGs) have a dismal prognosis, where the maximum tolerated dose (MTD) of IV terameprocol (5 days/month), a transcriptional inhibitor of specificity protein 1 (Sp1)-regulated proteins, is 1,700 mg/day with median area under the plasma concentration-time curve (AUC) of 31.3 μg∗h/mL. Given potentially increased efficacy with sustained systemic exposure and challenging logistics of daily IV therapy, here we investigate oral terameprocol for rHGGs in a multicenter, phase 1 trial (GATOR). Using a 3 + 3 dose-escalation design, we enroll 20 patients, with median age 60 years (range 31-80), 70% male, and median one relapse (range 1-3). Fasting patients tolerate 1,200 mg/day (n = 3), 2,400 mg/day (n = 6), 3,600 mg/day (n = 3), and 6,000 mg/day (n = 2) oral doses without major toxicities. However, increased dosage does not lead to increased systemic exposure, including in fed state (6,000 mg/day, n = 4), with maximal AUC <5 μg∗h/mL. These findings warrant trials investigating approaches that provide sustained systemic levels of transcription inhibitors to exploit their therapeutic potential. This study was registered at ClinicalTrials.gov (NCT02575794)., Competing Interests: Declaration of interests M.S.A. – grants: Seagen, AstraZeneca, BMS, Bayer, Incyte, Pharmacyclics, Novocure, MimiVax, and Merck. Consultation fees: Bayer, Novocure, Kiyatec, Insightec, GSK, Xoft, Nuvation, Celularity, SDP Oncology, Apollomics, Prelude Therapeutics, Janssen, Tocagen, Voyager Therapeutics, ViewRay, Caris Life Sciences, Pyramid Biosciences, Varian Medical Systems, Cairn Therapeutics, AnHeart Therapeutics, Menarini Ricerche, Sumitomo Pharma Oncology, Autem therapeutics, GT Medical Technologies, Allovir, Equillium Bio., QV Bioelectronics, and Theraguix. Scientific Advisory Board memberships: Cairn Therapeutics, Pyramid Biosciences, Bugworks, and Modifi Biosciences. Data Safety and Monitoring Committee membership: VBI Vaccines. Stock shareholder: MimiVax, CytoDyn, Trisalus Lifesciences, and MedInnovate Advisors, LLC. D.M.P. – consulting or advisory role: Orbus Therapeutics, Sumitomo Dainippon Pharma Oncology, Inc., Stemline Therapeutics, and Novocure. Research funding: Pfizer (Inst), Novartis (Inst), NeOnc Technologies (Inst), Orbus Therapeutics (Inst), Bristol Myers Squibb (Inst), Genentech/Roche (Inst), Pharmacyclics (Inst), Bayer (Inst), Karyopharm Therapeutics (Inst), Apollomics (Inst), Vigeo Therapeutics (Inst), Global Coalition for Adaptive Research (Inst), MimiVax (Inst), Ono Pharmaceutical (Inst), and Mylan (Inst). Equity ownership/stock options: Pfizer (Pharmaceuticals) and Gilead (Pharmaceuticals). M.H. – Data Safety Monitoring Board member: Parexel and Advarra. Institutional research funding: Novartis and Vanquish. P.Y.W. – research support: AstraZeneca, Black Diamond, Bristol Meyers Squibb, Celgene, Chimerix, Eli Lily, Erasca, Genentech/Roche, Kazia, MediciNova, Merck, Novartis, Nuvation Bio, Servier, Vascular Biogenics, and VBI Vaccines. Advisory board/consultant: AstraZeneca, Black Diamond, Celularity, Chimerix, Day One Bio, Genenta, Glaxo Smith Kline, Merck, Mundipharma, Novartis, Novocure, Nuvation Bio, Prelude Therapeutics, Sapience, Servier, Sagimet, Vascular Biogenics, and VBI Vaccines. R.C.H. has patents related to the drug terameprocol and is affiliated with Erimos Pharmaceuticals, which holds rights to the drug. S.G. is an independent contractor/consultant to Erimos Pharmaceuticals, LLC with no ownership or other financial interest in Erimos Pharmaceuticals or its products., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Hypotensive drugs mitigate the high-sodium diet-induced pro-inflammatory activation of mouse macrophages in vivo.
- Author
-
Cieślik M, Strobel SD, Bryniarski P, Twardowska H, Chmielowski A, Rudek M, Felkle D, Zięba K, Kaleta K, Jarczyński M, Nowak B, Bryniarski K, and Nazimek K
- Subjects
- Animals, Mice, Inflammation drug therapy, Macrophage Activation drug effects, Hypertension chemically induced, Hypertension drug therapy, Hypertension immunology, Male, Cytokines metabolism, Phagocytosis drug effects, Sodium, Dietary adverse effects, Inflammation Mediators metabolism, Antihypertensive Agents pharmacology, Macrophages drug effects, Macrophages metabolism, Macrophages immunology
- Abstract
Nowadays, there is an increasing emphasis on the need to alleviate the chronic inflammatory response to effectively treat hypertension. However, there are still gaps in our understanding on how to achieve this. Therefore, research on interaction of antihypertensive drugs with the immune system is extremely interesting, since their therapeutic effect could partly result from amelioration of hypertension-related inflammation, in which macrophages seem to play a pivotal role. Thus, current comprehensive studies have investigated the impact of repeatedly administered hypotensive drugs (captopril, olmesartan, propranolol, carvedilol, amlodipine, verapamil) on macrophage functions in the innate and adaptive immunity, as well as if drug-induced effects are affected by a high-sodium diet (HSD), one of the key environmental risk factors of hypertension. Although the assayed medications increased the generation of reactive oxygen and nitrogen intermediates by macrophages from standard fed donors, they reversed HSD-induced enhancing effects on macrophage oxidative burst and secretion of pro-inflammatory cytokines. On the other hand, some drugs increased macrophage phagocytic activity and the expression of surface markers involved in antigen presentation, which translated into enhanced macrophage ability to activate B cells for antibody production. Moreover, the assayed medications augmented macrophage function and the effector phase of contact hypersensitivity reaction, but suppressed the sensitization phase of cell-mediated hypersensitivity under HSD conditions. Our current findings contribute to the recognition of mechanisms, by which excessive sodium intake affects macrophage immune activity in hypertensive individuals, and provide evidence that the assayed medications mitigate most of the HSD-induced adverse effects, suggesting their additional protective therapeutic activity., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Occurrence of Dermacentor reticulatus in central-southern Poland, and potential threats to human and animal health.
- Author
-
Buczek A, Buczek W, Rudek M, Asman M, Świsłocka M, and Bartosik K
- Subjects
- Dogs, Animals, Male, Female, Humans, Poland epidemiology, Dermacentor microbiology, Rhipicephalus sanguineus, Rickettsia genetics
- Abstract
Introduction and Objective: Dermacentor reticulatus is one of the tick species of the greatest epidemiological importance in Europe. To date, the Eastern European and Western European populations of this tick species have been separated by an area located in Poland where the species has never been found. In this study, newly discovered D. reticulatus localities in areas transformed by human activities in central-southern Poland are described., Material and Methods: The specimens of the ornate dog tick were identified among ticks collected from companion animals in 2010, 2012, 2013, and 2014. They were examined using PCR methods to detect Borrelia burgdorferi s.l., Rickettsia spp., Anaplasma phagocytophilum , Bartonella spp., Babesia spp., and Toxoplasma gondii . In the case of the positive results, the amplicons were sequenced and examined by a BLAST search., Results: In total, 6 specimens of D. reticulatus were collected (3 females and 3 males). As declared by the owners, animal hosts stayed in the same area throughout the study period and had never travelled outside their place of residence. As many as 3/6 (50%) of D. reticulatus adults removed from dogs were infected with Rickettsia raoultii ., Conclusions: The results expand the available data on the spread of the ornate dog tick and indicate that, since 2010, this tick species and Rickettsia raoultii transmitted by this tick species have probably been present in this area, which has a strongly transformed agricultural structure and and had previously been regarded as a D. reticulatus -free zone. The presence of the ornate dog tick in urban and suburban habitats in central-southern Poland poses new threats to the health of companion animals and humans associated with the transmission of pathogens by this species.
- Published
- 2024
- Full Text
- View/download PDF
8. Dynamics of torque teno virus load in kidney transplant recipients with indication biopsy and therapeutic modifications of immunosuppression.
- Author
-
Reineke M, Morath C, Speer C, Rudek M, Bundschuh C, Klein JAF, Mahler CF, Kälble F, Nusshag C, Beimler J, Zeier M, Bartenschlager R, Schnitzler P, and Benning L
- Abstract
Following kidney transplantation, lifelong immunosuppressive therapy is essential to prevent graft rejection. On the downside, immunosuppression increases the risk of severe infections, a major cause of death among kidney transplant recipients (KTRs). To improve post-transplant outcomes, adequate immunosuppressive therapy is therefore a challenging but vital aspect of clinical practice. Torque teno virus load (TTVL) was shown to reflect immune competence in KTRs, with low TTVL linked to an elevated risk for rejections and high TTVL associated with infections in the first year post-transplantation. Yet, little is known about the dynamics of TTVL after the first year following transplantation and how TTVL changes with respect to short-term modifications in immunosuppressive therapy. Therefore, we quantified TTVL in 106 KTRs with 108 clinically indicated biopsies, including 65 biopsies performed >12 months post-transplantation, and correlated TTVL to histopathology. In addition, TTVL was quantified at 7, 30, and 90 days post-biopsy to evaluate how TTVL was affected by changes in immunosuppression resulting from interventions based on histopathological reporting. TTVL was highest in patients biopsied between 1 and 12 months post-transplantation (N = 23, median 2.98 × 10
7 c/mL) compared with those biopsied within 30 days (N = 20, median 7.35 × 103 c/mL) and > 1 year post-transplantation (N = 65, median 1.41 × 104 c/mL; p < 0.001 for both). Patients with BK virus-associated nephropathy (BKVAN) had significantly higher TTVL than patients with rejection ( p < 0.01) or other pathologies ( p < 0.001). When converted from mycophenolic acid to a mTOR inhibitor following the diagnosis of BKVAN, TTVL decreased significantly between biopsy and 30 and 90 days post-biopsy ( p < 0.01 for both). In KTR with high-dose corticosteroid pulse therapy for rejection, TTVL increased significantly between biopsy and 30 and 90 days post-biopsy ( p < 0.05 and p < 0.01, respectively). Of note, no significant changes were seen in TTVL within 7 days of changes in immunosuppressive therapy. Additionally, TTVL varied considerably with time since transplantation and among individuals, with a significant influence of age and BMI on TTVL ( p < 0.05 for all). In conclusion, our findings indicate that TTVL reflects changes in immunosuppressive therapy, even in the later stages of post-transplantation. To guide immunosuppressive therapy based on TTVL, one should consider inter- and intraindividual variations, as well as potential confounding factors., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Reineke, Morath, Speer, Rudek, Bundschuh, Klein, Mahler, Kälble, Nusshag, Beimler, Zeier, Bartenschlager, Schnitzler and Benning.)- Published
- 2024
- Full Text
- View/download PDF
9. Induction of double-strand breaks with the non-steroidal androgen receptor ligand flutamide in patients on androgen suppression: a study protocol for a randomized, double-blind prospective trial.
- Author
-
Lee E, Coulter J, Mishra A, Caramella-Pereira F, Demarzo A, Rudek M, Hu C, Han M, DeWeese TL, Yegnasubramanian S, and Song DY
- Subjects
- Male, Humans, Androgens, Androgen Antagonists therapeutic use, Receptors, Androgen, Ligands, Prospective Studies, Treatment Outcome, DNA, Randomized Controlled Trials as Topic, Flutamide therapeutic use, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology
- Abstract
Background: Prostate cancer remains the most prevalent malignancy and the second-leading cause of cancer-related death in men in the USA. Radiation therapy, typically with androgen suppression, remains a mainstay in the treatment of intermediate- and high-risk, potentially lethal prostate cancers. However, local recurrence and treatment failure remain common. Basic and translational research has determined the potential for using androgen receptor (AR) ligands (e.g., dihydrotestosterone and flutamide) in the context of androgen-deprived prostate cancer to induce AR- and TOP2B-mediated DNA double-strand breaks (DSBs) and thereby synergistically enhance the effect of radiation therapy (RT). The primary aim of this study is to carry out pharmacodynamic translation of these findings to humans., Methods: Patients with newly diagnosed, biopsy-confirmed localized prostatic adenocarcinoma will be recruited. Flutamide, an oral non-steroidal androgen receptor ligand, will be administered orally 6-12 h prior to prostate biopsy (performed under anesthesia prior to brachytherapy seed implantation). Key study parameters will include the assessment of DNA double-strand breaks by γH2A.x foci and AR localization to the nucleus. The initial 6 patients will be treated in a single-arm run-in phase to assess futility by establishing whether at least 2 subjects from this group develop γH2A.x foci in prostate cancer cells. If this criterion is met, the study will advance to a two-arm, randomized controlled phase in which 24 participants will be randomized 2:1 to either flutamide intervention or placebo standard-of-care (with all patients receiving definitive brachytherapy). The key pharmacodynamic endpoint will be to assess whether the extent of γH2A.x foci (proportion of cancer cells positive and number of foci per cancer cell) is greater in patients receiving flutamide versus placebo. Secondary outcomes of this study include an optional, exploratory analysis that will (a) describe cancer-specific methylation patterns of cell-free DNA in plasma and urine and (b) assess the utility of serum and urine samples as a DNA-based biomarker for tracking therapeutic response., Discussion: This study will confirm in humans the pharmacodynamic effect of AR ligands to induce transient double-strand breaks when administered in the context of androgen deprivation as a novel therapy for prostate cancer. The findings of this study will permit the development of a larger trial evaluating flutamide pulsed-dose sequencing in association with fractionated external beam RT (+/- brachytherapy). The study is ongoing, and preliminary data collection and recruitment are underway; analysis has yet to be performed., Trial Registration: ClinicalTrials.gov NCT03507608. Prospectively registered on 25 April 2018., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
10. Nelfinavir Inhibition of Kaposi's sarcoma-associated herpesvirus protein expression and capsid assembly.
- Author
-
Li M, Smith B, Jaeyeun L, Petr J, Wiseman R, Anders N, Rudek M, Ambinder R, and Desai P
- Abstract
Background: Antiviral therapies that target herpesviruses are clinically important. Nelfinavir is a protease inhibitor that targets the human immunodeficiency virus (HIV) infections aspartyl protease. Previous studies demonstrated that this drug could also inhibit Kaposi's sarcoma-associated herpesvirus (KSHV) production. Our laboratory demonstrated nelfinavir can effectively inhibit herpes simplex virus type 1 (HSV-1) replication. For HSV-1 we were able to determine that virus capsids were assembled and exited the nucleus but did not mature in the cytoplasm indicating the drug inhibited secondary envelopment of virions., Methods: For KSHV, we recently derived a tractable cell culture system that allowed us to analyze the virus replication cycle in detail. We used this system to further define the stage at which nelfinavir inhibits KSHV replication., Results: We discovered that nelfinavir inhibits KSHV extracellular virus production. This was seen when the drug was incubated with the cells for 3 days and when we pulsed the cells with the drug for 1-5 minutes. When KSHV infected cells exposed to the drug were examined using ultrastructural methods there was an absence of mature capsids in the nucleus indicating a defect in capsid assembly. Because nelfinavir influences the integrated stress response (ISR), we examined the expression of viral proteins in the presence of the drug. We observed that the expression of many were significantly changed in the presence of drug. The accumulation of the capsid triplex protein ORF26 was markedly reduced. This is an essential protein required for herpesvirus capsid assembly., Conclusions: Our studies confirm that nelfinavir inhibits KSHV virion production by disrupting virus assembly and maturation. Of interest is that inhibition requires only a short exposure to drug. The source of infectious virus in saliva has not been defined in detail but may well be lymphocytes or other cells in the oral mucosa. Thus, it might be that a "swish and spit" exposure rather than systemic administration would prevent virion production., Competing Interests: Competing interests The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
11. Donor-Derived Cell-Free DNA (dd-cfDNA) in Kidney Transplant Recipients With Indication Biopsy-Results of a Prospective Single-Center Trial.
- Author
-
Benning L, Morath C, Fink A, Rudek M, Speer C, Kälble F, Nusshag C, Beimler J, Schwab C, Waldherr R, Zeier M, Süsal C, and Tran TH
- Subjects
- Humans, Biopsy, Graft Rejection diagnosis, Prospective Studies, Tissue Donors, Transplant Recipients, Cell-Free Nucleic Acids, Kidney Transplantation adverse effects
- Abstract
Donor-derived cell-free DNA (dd-cfDNA) identifies allograft injury and discriminates active rejection from no rejection. In this prospective study, 106 kidney transplant recipients with 108 clinically indicated biopsies were enrolled at Heidelberg University Hospital between November 2020 and December 2022 to validate the clinical value of dd-cfDNA in a cohort of German patients. dd-cfDNA was quantified at biopsy and correlated to histopathology. Additionally, dd-cfDNA was determined on days 7, 30, and 90 post-biopsy and analyzed for potential use to monitor response to anti-rejection treatment. dd-cfDNA levels were with a median (IQR) % of 2.00 (0.48-3.20) highest in patients with ABMR, followed by 0.92 (0.19-11.25) in patients with TCMR, 0.44 (0.20-1.10) in patients with borderline changes and 0.20 (0.11-0.53) in patients with no signs of rejection. The AUC for dd-cfDNA to discriminate any type of rejection including borderline changes from no rejection was at 0.72 (95% CI 0.62-0.83). In patients receiving anti-rejection treatment, dd-cfDNA levels significantly decreased during the 7, 30, and 90 days follow-up compared to levels at the time of biopsy ( p = 0.006, p = 0.002, and p < 0.001, respectively). In conclusion, dd-cfDNA significantly discriminates active rejection from no rejection. Decreasing dd-cfDNA following anti-rejection treatment may indicate response to therapy. Clinical Trial Registration : https://drks.de/search/de/trial/DRKS00023604, identifier DRKS00023604., Competing Interests: This study received funding from CareDx Inc. (Brisbane, CA). The funder had the following involvement with the study: funding for kits and supplies for dd-cfDNA testing., (Copyright © 2023 Benning, Morath, Fink, Rudek, Speer, Kälble, Nusshag, Beimler, Schwab, Waldherr, Zeier, Süsal and Tran.)
- Published
- 2023
- Full Text
- View/download PDF
12. Off-label, but on target: the evidence needed to implement alternative dosing regimens of anticancer drugs.
- Author
-
Overbeek JK, Ter Heine R, Verheul HMW, Chatelut E, Rudek MA, Gurney H, Plummer R, Gilbert DC, Buclin T, Burger DM, Bloemendal HJ, and van Erp NP
- Subjects
- Humans, Clinical Protocols, Off-Label Use, Antineoplastic Agents therapeutic use
- Published
- 2023
- Full Text
- View/download PDF
13. Safety and Tolerability of Carboplatin and Paclitaxel in Cancer Patients with HIV (AMC-078), an AIDS Malignancy Consortium (AMC) Study.
- Author
-
Haigentz M, Moore P, Bimali M, Cooley T, Sparano J, Rudek M, Ratner L, Henry D, Ramos J, Deeken J, Rubinstein P, and Chiao E
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Carboplatin adverse effects, Cytochrome P-450 CYP3A Inhibitors therapeutic use, Humans, Paclitaxel adverse effects, Acquired Immunodeficiency Syndrome chemically induced, Acquired Immunodeficiency Syndrome drug therapy, HIV Infections complications, HIV Infections drug therapy, Neoplasms complications, Neoplasms drug therapy
- Abstract
Background: Persons living with human immunodeficiency virus are an underserved population for evidence-based cancer treatment. Paclitaxel and carboplatin (PCb) is an active regimen against a variety of solid tumors, including several seen in excess in patients with HIV infection. We performed a pilot trial to evaluate the safety of full-dose PCb in people living with human immunodeficiency virus and cancer., Methods: Eligible patients, stratified by concurrent antiretroviral therapy (ART) that included CYP3A4 inhibitors or not, received paclitaxel (175 mg/m2) in combination with carboplatin (target AUC 6) intravenously every 3 weeks for up to 6 cycles., Results: Sixteen evaluable patients received 64 cycles of PCb, including 6 patients treated with CYP3A4 inhibiting ART (ritonavir). The adverse event profile was consistent with the known toxicity profile of PCb, with no differences between the 2 strata. There were 4 partial responses (25%, 95% CI: 7%-52%), and overall, CD4+ lymphocyte count was similar after completion of therapy (median: 310/μL) compared with baseline values (median: 389/μL). Pharmacokinetic studies in 6 patients revealed no significant differences in Cmax or AUCinf for paclitaxel between the 2 cohorts., Conclusion: Full doses of PCb chemotherapy are tolerable when given concurrently with ART in people living with human immunodeficiency virus with cancer, including patients receiving CYP3A4 inhibitors., Clinicaltrials.gov Identifier: NCT01249443., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.