241 results on '"P. Gajate"'
Search Results
202. En la guerra de África (1921).
- Author
-
Bajo, María Gajate
- Subjects
SPANISH history -- 1868-1931 ,NONFICTION - Abstract
A review of the book "En la guerra de África (1921)" edited by Luis Arias González is presented.
- Published
- 2010
203. Antimyeloma Efficacy of Plitidepsin (Aplidin®): From Bench to the Bedside.
- Author
-
Ocio, Enrique M., Mitsiades, Constantine, Mateos, M. Victoria, Maiso, Patricia, Mollinedo, Faustino, Garayoa, Mercedes, Gajate, Consuelo, Blade, Joan, Prosper, Felipe, Lahuerta, Juan José, Mitsiades, Nicholas, McMullan, Ciaran J., Munshi, Nikhil C., Hideshima, Teru, Chauhan, Dharminder, Cuevas, Carmen, Avilés, Pablo, Faircloth, Glynn, Richardson, Paul G., Pandiella, Atanasio, Anderson, Kenneth C., and Miguel, J.F. San
- Abstract
Introduction Plitidepsin is a cyclic depsipeptide isolated from the marine tunicate, Aplidium albicans with promising antitumor activity. This work represents a comprehensive study (in vitro, in vivo and clinical) of its antimyeloma efficacy. Material & Methods In vitro studies were performed in 23 multiple myeloma (MM) cell lines and in cells from 16 MM patients. For the in vivo analysis a human plasmocytoma model in CB17-SCID mouse was used. Mice were randomized to receive Aplidin® 100 μg/Kg ip x 7 days/week (n=9), Aplidin® 140 μg/Kg ip x 5 days/week (n=7) or vehicle alone (n=9). The clinical efficacy of Aplidin® in relapsed/refractory patients was evaluated in a non-randomized two-stage Phase II, multicenter, clinical trial. Dosage of Aplidin® was 5 mg/m2 every 2 weeks. Results Aplidin® showed clear in vitro efficacy (IC50:1–10 nM) in the 23 cell lines tested including those resistant to dexamethasone, melphalan or doxorubicin. It was also active in the presence of microenvironment (IL-6, IGF-1 and BMSCs). Thirteen out of the 16 patient samples were sensitive to Aplidin® with >80% cell death in 8 cases and 60–80% in the remaining ones without significant toxicity in non tumor cells. Combination of Aplidin® with dexamethasone, bortezomib or lenalidomide showed clear potentiation. Aplidin® acts by inducing apoptosis with caspase−3, −7, −8, −9 and PARP cleavage. It also involves the activation of p38 and JNK signalling, Fas/CD95 translocation to lipid rafts and downregulation of Mcl-1 and myc. In mice studies, both schedules of treatment reduced tumor growth and increased survival with statistical differences in the group receiving 140 μg/Kg x 5d/week (p=0.04, Log Rank p=0.02). No significant toxicity was observed. These data provided the rationale for a clinical trial that has included 31 patients with relapsed/refractory MM. Median age was 65 years (47–82) and the median number of prior lines of therapy was 4 (range: 1–9) including autologous stem cell transplant (60%), thalidomide (58%) and bortezomib (48%). Out of the 26 evaluable patients, 2 (8%) achieved PR and 3 (12%) MR. Eight patients (31%) remained in stable disease (SD). Due to the synergism with dexamethasone observed in the in vitro studies, the protocol was amended to allow the addition of this agent in pts progressing after 3 cycles or with SD after 4 cycles. With a median follow-up of 14 months (range: 6.8–16.3), the time to progression in responding pts was 5.8 months (4.9–7.6). The most common G3-4 adverse events were fatigue (7%), serum creatine phosphokinase increase (7%), muscle toxicity (10%) and hepatic toxicity (10%). No significant hematologic toxicity or neuropathy was observed. Conclusion Aplidin® is effective both as a single agent and in combination with dexamethasone in the in vitro and in vivo settings. Its activity in relapsed/refractory MM patients is promising with an acceptable toxicity profile.
- Published
- 2007
- Full Text
- View/download PDF
204. Potential Colonization of the Peridomicile by Triatoma guasayana (Hemiptera: Reduviidae) in Santiago del Estero, Argentina
- Author
-
Gajate, Patricia P., Bottazzi, MaríA V., Pietrokovsky, Silvia M., and Wisnivesky-Colli, Cristina
- Abstract
Sylvatic triatomines might use the peridomicile as a 1st step in the process of domiciliation. Therefore, we evaluated the capability of sylvatic species to colonize the peridomicile of a rural area in the Province of Santiago del Estero, Argentina. The research was carried out in 6 houses in the village of Trinidad. The person per hour capture method was employed to determine the presence of triatomines in all the buildings (n = 44). Dispersing adults were collected by means of light traps and by villagers when approaching their houses. Triatoma infestans (Klug) was the most abundant species followed by the sylvatic Triatoma guasayana Wygodzinsky & Abalos. The branch pens, which included cacti, Opuntia quimilo, and bromeliads in their structure, were significantly associated with T. guasayana. Most of these insects had fed on domestic blood sources. With the exception of 1 Triatoma sordida (Stål), dispersing adults were T. guasayana; among those approaching houses, 12 were females (2 of which were infected with Trypanosoma cruzi Chagas) and 3 were males. T. guasayana was found to be capable of intensively invading the intradomicile and the peridomicile, showing a high tendency to settle in the ecotopes which included nontransformed raw material from the wild and where T. infestans was less abundant.
- Published
- 1996
- Full Text
- View/download PDF
205. La construcción del mito de Franco.
- Author
-
Gajate Bajo, María
- Published
- 2011
206. José Chabás Bordehore (1877-1963). Tuberculosis y medicina social en la Valencia del primer tercio del siglo XX.
- Author
-
Gajate Bajo, María
- Abstract
The book "José Chabás Bordehore (1877-1963). Tuberculosis y medicina social en la Valencia del primer tercio del siglo XX" by Josep L. Barona is reviewed.
- Published
- 2008
207. Rogaratinib Plus Atezolizumab in Cisplatin-Ineligible Patients With FGFR RNA-Overexpressing Urothelial Cancer: The FORT-2 Phase 1b Nonrandomized Clinical Trial.
- Author
-
Sweis RF, Gajate P, Morales-Barrera R, Lee JL, Necchi A, de Braud F, Penel N, Grünwald V, Maruzzo M, Meran J, Ishida TC, Bao W, Zhou Y, Ellinghaus P, and Rosenberg JE
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Receptor, Fibroblast Growth Factor, Type 1 genetics, Aged, 80 and over, Urologic Neoplasms drug therapy, Urologic Neoplasms genetics, Urologic Neoplasms pathology, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell genetics, RNA, Messenger genetics, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms pathology, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cisplatin administration & dosage, Cisplatin therapeutic use, Cisplatin adverse effects, Receptor, Fibroblast Growth Factor, Type 3 genetics
- Abstract
Importance: The oral pan-fibroblast growth factor receptor inhibitor rogaratinib previously demonstrated encouraging safety and efficacy in a phase 1 study of patients with urothelial cancer (UC) overexpressing FGFR messenger RNA (mRNA)., Objective: To evaluate the safety, pharmacokinetics, and preliminary efficacy of rogaratinib in combination with the programmed cell death 1 ligand 1 (PD-L1) inhibitor atezolizumab in cisplatin-ineligible patients with FGFR mRNA-positive, locally advanced/metastatic UC., Design Setting and Participants: The FORT-2 nonrandomized clinical trial was an open-label, single-arm, multicenter study conducted between May 15, 2018, and July 16, 2021, in 30 centers across Asia, Europe, and North America. Eligible patients had locally advanced/metastatic UC with FGFR1 / 3 mRNA overexpression and were ineligible for cisplatin-based chemotherapy. The data analysis was completed from July 2022 to September 2022., Interventions: Patients received rogaratinib 600 mg or rogaratinib 800 mg twice daily in combination with intravenous atezolizumab 1200 mg every 21 days., Main Outcomes and Measures: Primary end points included safety, tolerability, and the recommended phase 2 dose (RP2D) of rogaratinib in combination with atezolizumab., Results: Among 153 patients screened, 73 (48%) had tumors with FGFR1/3 mRNA overexpression, and 37 patients were enrolled and treated (median [range] age, 75.0 [47.0-85.0] years; 32 [87%] male). The most common treatment-emergent adverse events (TEAEs) included diarrhea in 23 patients (62%), hyperphosphatemia in 19 (51%), and fatigue in 15 (41%). Grade 3 or higher TEAEs were reported in 27 patients (73%), and 4 grade 5 TEAEs were reported, though unrelated to treatment. The RP2D was rogaratinib 600 mg in combination with atezolizumab 1200 mg. At the RP2D, the overall response rate was 53.8% in the rogaratinib 600 mg group, including 4 patients (15%) with complete responses; 12 responders (86%) did not have an FGFR3 gene alteration, and 11 (79%) had low PD-L1 expression., Conclusions and Relevance: In this phase 1b nonrandomized clinical trial, rogaratinib plus atezolizumab demonstrated a manageable safety profile, with no unexpected safety signals. Efficacy for this combination at the RP2D was observed in tumors with low PD-L1 and was not dependent on FGFR3 gene alterations, suggesting broad potential benefit for patients with locally advanced/metastatic UC and FGFR mRNA overexpression., Trial Registration: ClinicalTrials.gov Identifier: NCT03473756., Competing Interests: Conflict of Interest Disclosures: Dr Sweis reported grants paid to institution from Ascendis, AstraZeneca, Astellas, ALX Oncology, Bayer, BMS, CytomX, Eisai, Genentech/Roche, Gilead, Immunocore, Jounce, Loxo Oncology, Lilly, Merck, Moderna, Mirati, Novartis, Pfizer, Pionyr, Pyxis, QED, Regeneron, and Scholar Rock outside the submitted work; personal fees from AbbVie, Aveo, Eisai, Exelixis, Gilead, Janssen, and Lilly outside the submitted work; and a patent pending for Neoantigens in Cancer (PCT/US2020/031357). Dr Gajate reported personal fees from BMS, Merck, Roche, Pfizer, Astellas, Janssen, and MSD outside the submitted work. Dr Morales-Barrera reported serving in an advisory role for MSD, Pfizer, Merck, Janssen, and Astellas, as well as receiving honoraria or travel expenses from Roche, Sanofi Aventis, Astellas, Janssen, MSD, Bayer, Merck, and Pfizer. Dr de Braud reported personal fees from Pierre Fabre, Mattioli 1885, MSD, IQVIA, BMS, Indena, Incyte, Taiho, Menarini, Novartis, Roche, Sanofi, AccMed, Itanet, ESO, Dephaforum, Nadirex, Events, Fare Comunicazione, Motore Sanità, Effetti, Ambrosetti, Dynamicom Education, and AstraZeneca outside the submitted work. Dr Grünwald reported personal fees from Bristol Myers Squibb, Merck Serono, MSD, AstraZeneca, and Astellas during the conduct of the study; personal fees from AstraZeneca, Gilead, Pfizer, Novartis/AAA, Janssen-Cilag, Amgen, Ipsen, Eisai, Debiopharm, Apogepha, PCI Biotech, Synthekine, and Oncorena outside the submitted work. Dr Rosenberg reported personal fees from Bayer during the conduct of the study; personal fees from Janssen, AstraZeneca, Chugai, Merck, Seagen, Pfizer, Astellas, Genentech/Roche, EMD-Serono, Boehringer Ingelheim, Lilly/Loxo Oncology, Tyra Biosciences, QED Therapeutics, Gilead, Hengrui, Century Therapeutics, and Aktis Consultant outside the submitted work. No other disclosures were reported., (Copyright 2024 Sweis RF et al. JAMA Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
208. Detrimental effect of an early exposure to antibiotics on the outcomes of immunotherapy in a multi-tumor cohort of patients.
- Author
-
Guerrero P, Albarrán V, González-Merino C, García de Quevedo C, Sotoca P, Chamorro J, Rosero DI, Barrill A, Alía V, Calvo JC, Moreno J, Pérez de Aguado P, Álvarez-Ballesteros P, San Román M, Serrano JJ, Soria A, Olmedo ME, Saavedra C, Cortés A, Gómez A, Lage Y, Ruiz Á, Ferreiro MR, Longo F, Guerra E, Martínez-Delfrade Í, Garrido P, and Gajate P
- Abstract
Background: Immune checkpoint inhibitors (ICI) have changed the therapeutic landscape of many solid tumors. Modulation of the intestinal microbiota by antibiotics (Abx) has been suggested to impact on ICI outcomes., Methods: Retrospective analysis of 475 patients with advanced solid tumors treated with ICI from 2015 to 2022. For each patient, the use of Abx was recorded from 1 month before ICI initiation until disease progression or death. The impact of Abx on objective response rates (ORR), disease control rates (DCR), progression-free survival (PFS), and overall survival (OS) was analyzed. Kaplan-Meier and log-rank tests were used to compare survival outcomes., Results: In total 475 patients with advanced solid tumors were evaluated. Median age was 67.5 years and performance status (PS) was 0-1 in 84.6%. 66.5% of patients received Abx during treatment with ICI, mainly beta-lactams (53.8%) and quinolones (35.9%). The early exposure to Abx (from 60 days before to 42 days after the first cycle of ICI) was associated with a lower ORR (27.4% vs 39.4%; P < .01), a lower DCR (37.3% vs 57.4%; P < .001), lower PFS (16.8 m vs 27.8 m; HR 0.66; P < .001]) and lower OS (2.5 m vs 6.6 m; HR 0.68; P = .001]). The negative impact of Abx on OS and PFS was confirmed by a multivariable analysis. This effect was not observed among patients receiving Abx after 6 weeks from ICI initiation., Conclusions: Our results validate the hypothesis of a detrimental effect of an early exposure to Abxon the efficacy of ICI in a multi-tumor cohort of patients., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
209. Nomograms to Appraise The Risk of Chronic Kidney Disease After Radical Cystectomy: Shifting The Focus to Prevention.
- Author
-
Artiles Medina A, Mínguez Ojeda C, Subiela Henríquez JD, Muriel García A, Sánchez González Á, Mata Alcaraz M, Brasero Burgos J, Gajate Borau P, Gómez Dos Santos V, Jiménez Cidre MÁ, and Burgos Revilla FJ
- Abstract
Introduction: Our objectives were to analyse the incidence of changes in renal function after radical cystectomy (RC) and determine the factors responsible for those changes, as a basis for rethinking strategies to ensure early detection and development of a risk-adapted approach., Patients and Methods: A single-centre retrospective study included 316 patients who underwent RC between 2010 and 2019. A competing risk Cox model, whereby death from any cause was treated as a censoring event, was used to establish nomograms to analyze the prognostic factors for CKD at 2 and 5 years. The nomograms were validated based on discrimination using the C-index, calibration plots and analysis of net benefit from decision curves., Results: During a median follow-up of 48.73 months (0.13-156.67), 138 patients (43.7%) developed CKD. The probability of CKD development at 2 and 5 years was 41.3% (95% CI, 35.8-47.2) and 48.5% (95% CI, 42.8-54.6), respectively. Hypertension (HR 1.69, 95% CI, 1.23-2.34), prior hydronephrosis (HR 1.62, 95% CI, 1.17-2.25), acute kidney injury (AKI) during the immediate postoperative period (HR 1.88, 95% CI, 1.35-2.61) and readmission due to urinary tract infection (HR 1.41, 95% CI, 1.01-1.96) were predictors of 2-year CKD. Hydronephrosis at follow-up computed tomography (HR 2.21, 95% CI, 1.60-3.07), prior hydronephrosis (HR 1.54, 95% CI, 1.09-2.15), AKI during the immediate postoperative period (HR 1.77, 95% CI, 1.27-2.46) and hypertension (HR 1.60, 95% CI, 1.16-2.21) were predictors for 5-year CKD. Prior eGFR ≥ 90 mL/min/1.73 m
2 was a protective factor (HR 0.50, 95% CI, 0.32-0.80 and HR 0.48, 95% CI, 0.30-0.78 for 2- and 5-year CKD, respectively). The resulting nomograms were based on these prognostic factors., Conclusion: Almost half of the patients had developed CKD at 5 years. Thus, it is crucial to identify patients at risk of developing CKD in order to initiate renal function-sparing measures and tailor follow-up protocols. The proposed nomograms effectively predicted CKD in these patients., Competing Interests: Disclosure The authors declare no conflicts of interest in association with the present study., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
210. Is trimodal therapy the current standard for muscle-invasive bladder cancer?
- Author
-
López Valcárcel M, Barrado Los Arcos M, Ferri Molina M, Cienfuegos Belmonte I, Duque Santana V, Gajate Borau P, Fernández Ibiza J, Álvarez Maestro M, Sargos P, López Campos F, and Couñago F
- Subjects
- Humans, Combined Modality Therapy, Organ Sparing Treatments, Cystectomy methods, Urinary Bladder Neoplasms therapy, Urinary Bladder Neoplasms pathology, Neoplasm Invasiveness
- Abstract
Objective: The aim of this review is to summarize the current evidence and future perspectives of bladder-sparing treatment for MIBC., Methods: A non-systematic literature search in Medline/Pubmed was performed in October 2023 with the following keywords "bladder cancer", "bladder-sparing", "trimodal therapy", "chemoradiation", "biomarkers", "immunotherapy", "neoadjuvant chemotherapy", "radiotherapy"., Results: Urology guidelines recommend radical cystectomy as the standard curative treatment for muscle-invasive urothelial bladder cancer, reserving radiotherapy for patients who are unfit or who want to preserve their bladder. Given the morbidity and mortality of cystectomy and its impact on quality of life and bladder function, modern oncologic therapies are increasingly oriented toward organ preservation and maximizing functional outcomes while maintaining treatment efficacy. Trimodal therapy, which incorporates maximal transurethral resection followed by radiotherapy with concurrent radiosensitizing chemotherapy, is an effective regimen for bladder function preservation in well-selected patients. Despite the absence of comparative data from randomized trials, the two approaches seem to provide comparable oncologic outcomes. Studies are evaluating the expansion of eligibility criteria for trimodal therapy, the optimization of radiotherapy and immunotherapy delivery to further improve outcomes, and the validation of biomarkers to guide bladder preservation., Conclusions: Trimodal therapy has shown acceptable outcomes for bladder preservation; therefore, it provides a valid treatment option in well-selected patients., (Copyright © 2024 AEU. Published by Elsevier España, S.L.U. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
211. Clinical Usefulness of Prostate-specific Membrane Antigen-ligand Positron Emission Tomography/Computed Tomography for the Detection of Prostate Cancer Biochemical Recurrence after Primary Radiation Therapy in Patients with Prostate-specific Antigen Below the Phoenix Threshold: Systematic Review and Meta-analysis.
- Author
-
Subiela JD, Gomis Sellés E, Maldonado A, Lopez Campos F, Aumatell Ovide J, Ajuria Illarramendi O, González-Padilla DA, Gajate P, Ortega Polledo LE, Alonso Y Gregorio S, Guerrero-Ramos F, Gómez Dos Santos V, Rodríguez-Patrón R, Calais J, Kishan AU, Burgos Revilla FJ, and Couñago F
- Subjects
- Male, Humans, Positron Emission Tomography Computed Tomography methods, Prostate pathology, Ligands, Neoplasm Recurrence, Local diagnostic imaging, Neoplasm Recurrence, Local pathology, Retrospective Studies, Prostate-Specific Antigen, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms radiotherapy, Prostatic Neoplasms pathology
- Abstract
Aims: After primary radiotherapy, biochemical recurrence is defined according to the Phoenix criteria as a prostate-specific antigen (PSA) value >2 ng/ml relative to the nadir. Several studies have shown that prostate-specific membrane antigen (PSMA)-ligand positron emission tomography/computed tomography (PET/CT) can help in detecting recurrence in patients with low PSA values. This study aimed to assess the detection rate and patterns of PSMA-ligand PET/CT uptake in patients with suspected biochemical recurrence after primary radiotherapy and with PSA levels below the Phoenix threshold., Materials and Methods: The meta-analysis was carried out in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement. Articles providing data on patients with suspected prostate cancer recurrence after primary radiotherapy with a PSA value below the Phoenix threshold and who underwent PSMA-ligand PET/CT were included. Quality assessment was carried out using the Quality Assessment of Diagnostic Accuracy Studies-2 tool (QUADAS-2)., Results: In total, five studies were included, recruiting 909 patients (202 with PSA ≤2 ng/ml). The PSMA-ligand detection rate in the patients with ≤2 ng/ml ranged from 66 to 83%. The most frequent source of PSMA-ligand PET/CT uptake was local recurrence, followed by lymph node metastasis and bone metastasis. PSMA-ligand PET/CT uptake due to local-only recurrence was more likely in patients with PSA ≤2 ng/ml compared with PSA > 2 ng/ml: risk ratio 0.72 (95% confidence interval 0.58-0.89), P = 0.003. No significant differences were observed in the detection of PSMA-ligand uptake in other areas. Limitations include a lack of biopsy confirmation, cohort reports with small sample sizes and a potentially high risk of bias., Conclusion: A significant detection of PSMA-ligand-avid disease was observed in patients with PSA levels below the Phoenix threshold. There was a higher likelihood of detecting local-only uptake when the PSA value was ≤2 ng/ml. The findings suggest that a critical review of the Phoenix criteria may be warranted in the era of PSMA-ligand PET/CT and highlight the need for further prospective trials., (Copyright © 2023. Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
212. Stereotactic body radiation therapy for kidney cancer. Where do we stand?
- Author
-
Sosa-Fajardo P, Blanco-Suarez JM, Pineda-Munguía Á, Rubí-Olea L, Peleteiro-Higuero P, Gajate P, Zafra-Martín J, Siva S, Bossi A, López-Campos F, and Couñago F
- Subjects
- Humans, Radiosurgery adverse effects, Radiosurgery methods, Carcinoma, Renal Cell radiotherapy, Carcinoma, Renal Cell surgery, Kidney Neoplasms radiotherapy, Kidney Neoplasms surgery
- Abstract
At present, surgery is still the gold standard for the local treatment of renal cancer. Nonetheless, in several clinical scenarios, stereotactic body radiation therapy (SBRT) also known as stereotactic ablative body radiotherapy (SABR) is emerging as a highly effective ablative technique in fragile patients and those with significant comorbidities, as well as in cases where percutaneous therapy (cryoablation or radiofrequency) is not viable. However, considering the intrinsic radioresistance of renal tumors, the optimal treatment schemes have not been established. In oligometastatic patients, it has been reported that the control of the oligometastases can be a potentially curable approach. Being a technique than can be administered exclusively or in combination with systemic therapy, treatment individualization based on patient characteristics is key. Another scenario under investigation is oligoprogression, where SBRT offers the possibility of delaying further lines of systemic therapy by eliminating subclones of resistant tumor with ablative doses, with the additional opportunity of stimulating the immune system (immunomodulatory role). In this review, we have conducted an analysis of recently published studies that test the role of this technique in different clinical scenarios of this disease. We have found promising results that make SBRT a potent therapeutic approach with low toxicity. We also comment on ongoing studies that will generate the necessary evidence needed for the implementation of this technique in our daily clinical practice., (© 2023 The Japanese Urological Association.)
- Published
- 2023
- Full Text
- View/download PDF
213. Oncological and Renal Function Outcomes in Patients Who Underwent Simultaneous Radical Cystectomy and Nephroureterectomy for Synchronous or Metachronous Panurothelial Carcinoma.
- Author
-
Subiela JD, González-Padilla DA, Huguet J, Aumatell J, Rodríguez-Faba O, Krajewski W, Feliu AH, Mínguez C, Plaza JL, Artiles Medina A, Gajate P, Jiménez Cidre MÁ, Burgos Revilla J, Breda A, and Palou J
- Subjects
- Humans, Nephroureterectomy adverse effects, Cystectomy adverse effects, Retrospective Studies, Kidney pathology, Treatment Outcome, Carcinoma, Transitional Cell pathology, Urinary Bladder Neoplasms pathology
- Abstract
Objective: To assess clinical outcomes of patients who underwent simultaneous radical cystectomy (RC) and radical nephroureterectomy (RNU) for panurothelial carcinoma (PanUC)., Materials and Methods: A retrospective analysis of 67 patients who underwent simultaneous RC and unilateral RNU for PanUC, from 1996 to 2017. Kaplan-Meier estimates for remnant urothelium recurrence-free survival, metastasis-free survival, overall survival (OS), and cancer-specific survival (CSS) were performed. Cox multivariate models were constructed., Results: The median follow-up was 38 months, 29.8% of patients had a recurrence, 34.3% had metastasis, 67.2% of patients died from any cause, and 37.3% died from urothelial carcinoma. Overall survival and CSS rates at 5 years were 44% and 61%, respectively. In multivariate analysis, progression to muscle-invasive bladder cancer before surgery, presence of muscle-invasive stages at RC and/or RNU, and prostatic urethra involvement were predictors for worse metastasis-free survival and CSS. Forty-one patients (61.2%) had an estimated glomerular filtration rate (eGFR) <60 mL/min before surgery and the number rose to 56 (83.5%) after surgery; 29.8% patients needed renal function replacement therapy after surgery (16 haemodialysis and 4 renal transplant)., Conclusion: Patients with PanUC who undergo simultaneous surgery have adverse oncological (only 4 out of every 10 remain alive at 5 years) and functional outcomes (1 out of 3 will need renal function replacement therapy after surgery). Up to a third of the patients had a recurrence (urethra or contralateral kidney) within 18 months, justifying close surveillance or considering prophylactic urethrectomy. These data should help in counsel on morbidity and life expectancy., Competing Interests: Conflict of interest None., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
214. Current and Future Landscape of Perioperative Treatment for Muscle-Invasive Bladder Cancer.
- Author
-
Esteban-Villarrubia J, Torres-Jiménez J, Bueno-Bravo C, García-Mondaray R, Subiela JD, and Gajate P
- Abstract
Cisplatin-based neoadjuvant chemotherapy followed by radical cystectomy is the current standard of care for muscle-invasive bladder cancer (MIBC). However, less than half of patients are candidates for this treatment, and 50% will develop metastatic disease. Adjuvant chemotherapy could be offered if neoadjuvant treatment has not been administered for suitable patients. It is important to reduce the risk of systemic recurrence and improve the prognosis of localized MIBC. Systemic therapy for metastatic urothelial carcinoma has evolved in recent years. Immune checkpoint inhibitors and targeted agents, such as antibody-drug conjugates or FGFR inhibitors, are new therapeutic alternatives and have shown their benefit in advanced disease. Currently, several clinical trials are investigating the role of these drugs, as monotherapy and in combination with chemotherapy, in the neoadjuvant and adjuvant settings with promising outcomes. In addition, the development of predictive biomarkers could predict responses to neoadjuvant therapies.
- Published
- 2023
- Full Text
- View/download PDF
215. Her-2 Targeted Therapy in Advanced Urothelial Cancer: From Monoclonal Antibodies to Antibody-Drug Conjugates.
- Author
-
Albarrán V, Rosero DI, Chamorro J, Pozas J, San Román M, Barrill AM, Alía V, Sotoca P, Guerrero P, Calvo JC, Orejana I, Pérez de Aguado P, and Gajate P
- Subjects
- Humans, Antibodies, Monoclonal pharmacology, Tyrosine, Immunoconjugates therapeutic use, Immunoconjugates pharmacology, Carcinoma, Transitional Cell drug therapy, Antineoplastic Agents, Immunological therapeutic use, Urinary Bladder Neoplasms pathology
- Abstract
Metastatic urothelial cancer, associated with a poor prognosis, is still major cause of cancer-related death, with scarce options of effective treatment after progression to platinum-based chemotherapy and immunotherapy. The human epithelial growth factor receptor 2 (Her-2) has been identified as a new therapeutic target in medical oncology. However, despite the encouraging results in breast and gastric cancers, clinical trials with anti-Her-2 monoclonal antibodies and tyrosine-kinase inhibitors have shown limited efficacy of this strategy in urothelial tumors. Notably, more favorable data have been recently shown that antibody-drug conjugates are currently emerging as a novel promising approach for Her-2 targeted therapy in advanced urothelial cancer.
- Published
- 2022
- Full Text
- View/download PDF
216. A Randomized, Double-Blind Noninferiority Study to Evaluate the Efficacy of the Cabozantinib Tablet at 60 mg Per Day Compared with the Cabozantinib Capsule at 140 mg Per Day in Patients with Progressive, Metastatic Medullary Thyroid Cancer.
- Author
-
Capdevila J, Klochikhin A, Leboulleux S, Isaev P, Badiu C, Robinson B, Hughes BGM, Keam B, Parnis F, Elisei R, Gajate P, Gan HK, Kapiteijn E, Locati L, Mangeshkar M, Faoro L, Krajewska J, and Jarzab B
- Subjects
- Anilides adverse effects, Capsules therapeutic use, Humans, Protein Kinase Inhibitors therapeutic use, Pyridines, Tablets therapeutic use, Antineoplastic Agents therapeutic use, Carcinoma, Neuroendocrine pathology, Thyroid Neoplasms pathology
- Abstract
Background: Cabozantinib inhibits pathways involved in medullary thyroid cancer (MTC). Cabozantinib is approved as 140 mg/day in capsules for MTC and 60 mg/day in tablets for other solid tumors. This study compared the two doses in progressive metastatic MTC. Methods: In this Phase 4, randomized, double-blind noninferiority (NI) trial (NCT01896479), patients with progressive metastatic MTC were randomized 1:1 to cabozantinib 60 mg/day tablet or 140 mg/day capsules. The primary end point was progression-free survival (PFS) by blinded independent radiology committee (BIRC) per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. NI would be concluded if the upper 95% confidence interval [CI] for the PFS hazard ratio (HR) was less than the NI margin, 1.58. The secondary end point was objective response rate (ORR) by BIRC per RECIST v1.1; additional end points included safety and pharmacokinetics. Results: At data cutoff (July 15, 2020), 247 patients were randomized to the 60 mg/day tablet arm ( n = 123) and the 140 mg/day capsules arm ( n = 124). NI was not met (median PFS 11.0 months vs. 13.9 months in the 60 and 140 mg/day arms [HR 1.24; CI 0.90-1.70; p = 0.19]). The ORR was 33% in both arms. Generally, adverse event (AE) incidence was lower in the 60 mg/day arm (Grade 3/4, 63% vs. 72%), as were dose reductions (69% vs. 81%) and treatment discontinuations due to AEs (23% vs. 36%). Initially, cabozantinib plasma concentrations were higher in the 140 mg/day arm but became similar between arms at later time points. Conclusions: PFS NI of the cabozantinib 60 mg/day tablet vs. 140 mg/day capsules was not met. The 60 mg/day tablet had the same ORR and lower rates of AEs. Clinical Trial Registry: ClinicalTrials.gov NCT01896479.
- Published
- 2022
- Full Text
- View/download PDF
217. Limited T cell response to SARS-CoV-2 mRNA vaccine among patients with cancer receiving different cancer treatments.
- Author
-
Cortés A, Casado JL, Longo F, Serrano JJ, Saavedra C, Velasco H, Martin A, Chamorro J, Rosero D, Fernández M, Gion M, Martínez Jáñez N, Soria Rivas A, Alonso Gordoa T, Martínez Delfrade Í, Lage Y, López Miranda E, Olmedo ME, Reguera Puertas P, Gajate P, Molina Cerrillo J, Guerra Alia E, Fuentes Mateos R, Romero B, Rodríguez-Domínguez MJ, Vallejo A, and Carrato A
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Antibodies, Viral, COVID-19 Vaccines adverse effects, Humans, Prospective Studies, SARS-CoV-2, Vaccines, Synthetic, mRNA Vaccines, COVID-19 prevention & control, Neoplasms therapy
- Abstract
Introduction: Patients with cancer (PC) are at high risk of acquiring COVID-19 and can develop more serious complications. Deeper understanding of vaccines immunogenicity in this population is crucial for adequately planning vaccines programs. The ONCOVac study aimed to comprehensively assess the immunogenicity of mRNA-1273 vaccine in terms of humoral and cellular response., Methods: We conducted a prospective, single-center study including patients with solid tumours treated with cyclin-dependent kinases 4 and 6 inhibitors (CDK4/6i), immunotherapy (IT) or chemotherapy (CT). Patients were enrolled previously to vaccination with mRNA-1273. We also involved health care workers (HCW) to serve as a control group. We took blood samples before first dose administration (BL), after first dose (1D), and after second dose (2D). The primary objective was to compare the rate and magnitude of T cell response after second dose whereas safety and humoral response were defined as secondary objectives. We also collected patient reported outcomes after both the first and second vaccine dose and a six-month follow-up period to diagnose incident COVID-19 cases was planned., Results: The rate of specific anti-S serologic positivity (anti-S IgG cut-off point at 7,14 BAU/mL) was significantly higher in HCW compared to PC after 1D (100% versus 83.8%; p = 0.04), but similar after 2D (100% versus 95.8%; p = 0.5). This difference after 1D was driven by PC treated with CT (100% versus 64.5%; p = 0.001). Cellular response after 2D was significantly lower in PC than in HCW for both CD4+ (91.7% versus 59.7%; p = 0.001) and CD8+ (94.4% versus 55.6%; p < 0.001) T cells. We found a difference on pre-existing CD4+ T cell response in HCW comparing to PC (36% and 17%, p = 0.03); without difference in pre-existing CD8+ T cell response (31% and 23%, p = 0.5). After excluding patients with pre-existing T cell response, PC achieved even lower CD4+ (50.9% versus 95.5%, p < 0.001) and CD8+ (45.5% versus 95.5%, p < 0.001) T cell response compared with HCW. Regarding safety, PC reported notably more adverse events than HCW (96.6% versus 69.2%, p < 0.001)., Conclusion: We demonstrated that PC showed a similar humoral response but a lower T cell response following two doses of mRNA-1273 vaccination. Further studies are needed to complement our results and determine the implication of low T cell response on clinical protection of PC against COVID-19., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Alfonso Cortes declares the payment for presentations/speaker bureaus/manuscript-writing/educational events from GSK, AstraZeneca, Roche, MSD and Eisai. Alfonso Cortes declares consulting fees from Clovis, Lilly, Pfizer, GSK, Ferrer and Roche. Alfonso Cortes has received research grants from Pfizer. Alfonso Cortes declares support for attending meetings from Roche, Daiichi and Pfzer. Alfonso Cortes is co-founder of ONCARE. Jose Luis Casado and Federico Longo have not conflict of interest to declare. Juan José Serrano declares speaker bureau from Pierre-Fabre and travel/accomodation/expenses from Novartis. Cristina Saavedra declares travel/accomodation/expenses from Lilly and Pfizer. Hector Velasco, Alejandro Vallejo and Adrian San Miguel have not conflict of interest to declare. Jesus Chamorro and Diana Rosero have not conflict of interest to declare. María Fernandez, María Gion and Noelia Martinez have not conflict of interest to declare. Ainara Soria has not conflict of interest to declare. Teresa Alonso declares: Scientific Consultancy Role (speaker and advisory role) and travel grant from IPSEN, Pfizer, Bayer, Sanofi, Janssen, Astellas, Adacap, Eisai, Lilly, Novartis, BMS, Roche. Teresa Alonso declares the participation in Clinical trials from Roche, BMS, MSD, Pfizer, Novartis, IPSEN, Exelixis, Astrazeneca-Medimmune, Janssen, Lilly, Eisai, Astellas. Teresa Alonso declares research support from Roche, Pfizer, IPSEN. Iñigo Martinez and Yolanda Lage have not conflict of interest to declare. Elena López declares to receive advisory/consultancy honorarium from AstraZeneca, Pfizer, Roche, Novartis. Elena López has received speaker bureau/expert testimony honorarium from Roche, Novartis , Eisai, Astra Zeneca; and has received travel/accommodation/expenses from Roche, Novartis. María Eugenia Olmedo has not conflict of interest to declare. Pablo Reguera has not conflict of interest to declare. Pablo Gajate declares travel and educational support from BMS, MSD, Pfizer, Ipsen, Sanofi-Genzyne, Roche and Jansen. Pablo Gajate declares advisor and delivered lectures for BMS, MSD, Merck Serono, Pfizer, Ipsen, Roche, Adacap, Eisai, Sanofi-Genzyme, Novartis and Jansen. Javier Molina declares consultant, advisory or speaker roles for IPSEN, Roche, Pfizer, Sanofi, Janssen, and BMS. Javier Molina has received research grants from Pfizer, IPSEN and Roche. Eva Guerra declares: advisory/consultancy honorarium from AstraZeneca-MSD, Clovis Oncology, GSK-Tesaro, PharmaMar, Roche. Eva Guerra has received speaker bureau/expert testimony honorarium from AstraZeneca, PharmaMar, Roche, GSK; and has received travel/accommodation/expenses from Roche, TESARO, and Baxter. Raquel Fuentes, Beatriz Romero and Mario J Rodriguez-Dominguez have not conflict of interest. Alfredo Carrato has not conflict of interest to declare., (Copyright © 2022 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
218. Immunotherapy in Advanced Prostate Cancer: Current Knowledge and Future Directions.
- Author
-
López-Campos F, Gajate P, Romero-Laorden N, Zafra-Martín J, Juan M, Hernando Polo S, Conde Moreno A, and Couñago F
- Abstract
The advent of immunotherapy has revolutionized cancer treatment. Unfortunately, this has not been the case for metastatic castration-resistant prostate cancer (mCRPC), likely due to the heterogeneous and immune-suppressive microenvironment present in prostate cancer. The identification of molecular biomarkers that could predict response to immunotherapy represents one of the current challenges in this clinical scenario. The management of advanced castration-resistant prostate cancer is rapidly evolving and immunotherapy treatments, mostly consisting of immune checkpoint inhibitors combinations, BiTE
® (bispecific T-cell engager) immune therapies, and chimeric antigen receptors (CAR) are in development with promising results. This review analyses the current evidence of immunotherapy treatments for mCRPC, evaluating past failures and promising approaches and discussing the directions for future research.- Published
- 2022
- Full Text
- View/download PDF
219. Neurologic Toxicity of Immune Checkpoint Inhibitors: A Review of Literature.
- Author
-
Albarrán V, Chamorro J, Rosero DI, Saavedra C, Soria A, Carrato A, and Gajate P
- Abstract
Immune checkpoint inhibitors have entailed a change of paradigm in the management of multiple malignant diseases and are acquiring a key role in an increasing number of clinical sceneries. However, since their mechanism of action is not limited to the tumor microenvironment, their systemic activity may lead to a wide spectrum of immune-related side effects. Although neurological adverse events are much less frequent than gastrointestinal, hepatic, or lung toxicity, with an incidence of <5%, their potential severity and consequent interruptions to cancer treatment make them of particular importance. Despite them mainly implying peripheral neuropathies, immunotherapy has also been associated with an increased risk of encephalitis and paraneoplastic disorders affecting the central nervous system, often appearing in a clinical context where the appropriate diagnosis and early management of neuropsychiatric symptoms can be challenging. Although the pathogenesis of these complications is not fully understood yet, the blockade of tumoral inhibitory signals, and therefore the elicitation of cytotoxic T-cell-mediated response, seems to play a decisive role. The aim of this review was to summarize the current knowledge about the pathogenic mechanisms, clinical manifestations, and therapeutic recommendations regarding the main forms of neurotoxicity related to checkpoint inhibitors., Competing Interests: CS travel and educational support: Novartis, Lilly, Pfizer, MSD, BMS. AS travel and educational support: MSD, BMS, and Pierre Fabre and has served as advisor and delivered lectures for Novartis, MSD, BMS, Sanofi Aventis, Pierre Fabre, and Merck Serono. PG travel and educational support: Pfizer, Ipsen, Sanofi-Genzyne, Roche, and Jansen and has served as advisor and delivered lectures for BMS, MSD, Merck Serono, Pfizer, Ipsen, Roche, Adacap, Eisai, Sanofi-Genzyme, Novartis, and Jansen. The remaining 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., (Copyright © 2022 Albarrán, Chamorro, Rosero, Saavedra, Soria, Carrato and Gajate.)
- Published
- 2022
- Full Text
- View/download PDF
220. Surgery of pancreatic metastasis from renal cell carcinoma.
- Author
-
Vilar Tabanera A, Muñoz Muñoz P, Molina Villar JM, Gajate P, and Sanjuanbenito A
- Subjects
- Humans, Carcinoma, Renal Cell surgery, Kidney Neoplasms surgery, Pancreatic Neoplasms surgery
- Published
- 2022
- Full Text
- View/download PDF
221. Sunitinib and Evofosfamide (TH-302) in Systemic Treatment-Naïve Patients with Grade 1/2 Metastatic Pancreatic Neuroendocrine Tumors: The GETNE-1408 Trial.
- Author
-
Grande E, Rodriguez-Antona C, López C, Alonso-Gordoa T, Benavent M, Capdevila J, Teulé A, Custodio A, Sevilla I, Hernando J, Gajate P, Molina-Cerrillo J, Díez JJ, Santos M, Lanillos J, and García-Carbonero R
- Subjects
- Humans, Middle Aged, Nitroimidazoles, Phosphoramide Mustards, Progression-Free Survival, Sunitinib pharmacology, Sunitinib therapeutic use, Neoplasms, Second Primary, Pancreatic Neoplasms drug therapy
- Abstract
Background: Sunitinib (SUN)-induced hypoxia within the tumor could promote the activation of the prodrug evofosfamide (EVO), locally releasing the cytotoxic DNA alkylator bromo-isophosphoramide mustard. SUNEVO, a phase II, open-label, single-arm trial, investigated the potential synergy of SUN plus EVO in advanced progressive pancreatic neuroendocrine tumors (panNETs)., Methods: Systemic treatment-naïve patients with advanced or metastatic, unresectable, grade 1/2 panNETs with a Ki67 ≤20%, received EVO 340 mg/m
2 on days 8, 15, and 22 every 4 weeks and sunitinib 37.5 mg/day continuously. The primary endpoint was objective response rate, measured every 8 weeks by RECIST version 1.1., Results: From 2015 to 2018, 17 patients were enrolled. The median age was 62.4 years, 47% had a Ki67 >10%, and 70.6% had liver metastasis. Patients received a median of five and four cycles of SUN and EVO, respectively. After a median follow-up of 15.7 months, 17.6% of patients achieved a complete (n = 1) or partial response (n = 2), and 11 patients had stable disease (64.7%). The median progression-free survival was 10.4 months (95% confidence interval, 2.6-18.0). Treatment-related adverse events (grade ≥3) were observed in 64.7% of the patients, the most frequent being neutropenia (35.3%), fatigue (17.6%), and thrombopenia (11.8%). Treatment discontinuation due to toxicity was reported in 88.2% of the patients. No correlation was found between treatment response and DAXX, ATRX, MEN1, SETD2, and PTEN gene mutations., Conclusion: SUN plus EVO had a negative toxicity profile that should be taken into account for further clinical research in advanced panNETs. The combination showed moderate activity in terms of treatment response that did not correlate with somatic mutations. (Clinical trial identification number: NCT02402062) IMPLICATIONS FOR PRACTICE: Addition of hypoxia-activated prodrugs has been proposed as a potential mechanism to overcome tumor resistance to antiangiogenic agents. Sunitinib and evofosfamide, which were widely proposed as a potential synergistic option, showed modest efficacy in pancreatic neuroendocrine tumors (panNETs), reaching a median objective response rate of 17.6% and median progression-free survival of 10.4 months. Treatment response does not correlate with the biomarkers analyzed. The high systemic toxicity, with 88.2% of patients discontinuing the treatment, makes this therapeutic approach unfeasible and encourages future research to overcome panNETs' resistance to antiangiogenic agents with other therapies with a safer profile., (© 2021 AlphaMed Press.)- Published
- 2021
- Full Text
- View/download PDF
222. Clinical Practice Guidelines for Diagnosis and Management of Hypersensitivity Reactions to Quinolones.
- Author
-
Doña I, Blanca-López N, Boteanu C, Cueva-Oliver B, Fernández-Sánchez FJ, Gajate P, García-Avilés MC, García-Núñez I, Lobera T, Moreno E, Rojas P, and Rosado A
- Subjects
- Allergens immunology, Anti-Allergic Agents therapeutic use, Basophil Degranulation Test, Cross Reactions, Drug Hypersensitivity drug therapy, Drug Hypersensitivity etiology, Drug-Related Side Effects and Adverse Reactions drug therapy, Humans, Practice Guidelines as Topic, Quinolones therapeutic use, Skin Tests, Allergens adverse effects, Anti-Allergic Agents adverse effects, Desensitization, Immunologic methods, Drug Hypersensitivity diagnosis, Drug-Related Side Effects and Adverse Reactions diagnosis, Quinolones adverse effects
- Abstract
The consumption of quinolones as first-line treatment has increased in recent years, leading to an increase in the incidence of hypersensitivity reactions (HSRs) to this antibiotic group. Both diagnosis and management of HSRs to quinolones are complex and controversial. These practical guidelines aim to provide recommendations for effective clinical practice. The recommendations were drafted by an expert panel that reviewed the literature regarding HSRs to quinolones and analyzed controversies in this area. Most HSRs to quinolones are immediate and severe. The risk for HSRs is higher in patients who report allergy to ß-lactams, moxifloxacininduced anaphylaxis, and immediate reactions than in patients who report reactions to quinolones inducing other symptoms. The usefulness of skin tests in diagnosing HSRs to quinolones is controversial, with sensitivity and specificity varying between studies. Most in vitro tests are produced in-house, with no validated commercial options. The basophil activation test has proven useful for diagnosing immediate reactions, albeit with diverse results regarding sensitivity. Drug provocation testing is currently the gold standard for confirming or excluding the diagnosis and for finding safe alternatives, although it is contraindicated in patients with severe reactions. Cross-reactivity between quinolones has proven controversial in several studies, with the lowest cross-reactivity reported for levofloxacin. Desensitization may be considered in allergy to quinolones when no other alternatives are available.
- Published
- 2021
- Full Text
- View/download PDF
223. Lenvatinib in Patients With Advanced Grade 1/2 Pancreatic and Gastrointestinal Neuroendocrine Tumors: Results of the Phase II TALENT Trial (GETNE1509).
- Author
-
Capdevila J, Fazio N, Lopez C, Teulé A, Valle JW, Tafuto S, Custodio A, Reed N, Raderer M, Grande E, Garcia-Carbonero R, Jimenez-Fonseca P, Hernando J, Bongiovanni A, Spada F, Alonso V, Antonuzzo L, Spallanzani A, Berruti A, La Casta A, Sevilla I, Kump P, Giuffrida D, Merino X, Trejo L, Gajate P, Matos I, Lamarca A, and Ibrahim T
- Subjects
- Adult, Aged, Antineoplastic Agents adverse effects, Europe, Female, Gastrointestinal Neoplasms mortality, Gastrointestinal Neoplasms pathology, Humans, Male, Middle Aged, Neoplasm Grading, Neuroendocrine Tumors mortality, Neuroendocrine Tumors pathology, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Phenylurea Compounds adverse effects, Progression-Free Survival, Prospective Studies, Protein Kinase Inhibitors adverse effects, Quinolines adverse effects, Time Factors, Antineoplastic Agents therapeutic use, Gastrointestinal Neoplasms drug therapy, Neuroendocrine Tumors drug therapy, Pancreatic Neoplasms drug therapy, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors therapeutic use, Quinolines therapeutic use
- Abstract
Purpose: Approved systemic therapies for advanced gastroenteropancreatic neuroendocrine tumors (GEP-NETs) have shown limited capacity to reduce tumor burden and no antitumor activity after progression to targeted agents (TAs). We investigated the efficacy and safety of lenvatinib in patients with previously treated advanced GEP-NETs., Patients and Methods: This was a multicenter, single-arm, open-label, phase II trial with two parallel cohorts (ClinicalTrials.gov identifier: NCT02678780) involving 21 institutions in 4 European countries. Eligible patients had histologically confirmed advanced grade 1-2 pancreatic (panNET) or GI (GI-NET) NETs with documented tumor progression after treatment with a TA (panNET) or somatostatin analogs (GI-NET). Patients were treated with lenvatinib 24 mg once daily until disease progression or treatment intolerance. The primary end point was overall response rate by central radiology review. Secondary end points included progression-free survival, overall survival, duration of response, and safety., Results: Between September 2015 and March 2017, a total of 111 patients were enrolled, with 55 (panNET) and 56 (GI-NET) patients in each cohort. The median follow-up was 23 months. The overall response rate was 29.9% (95% CI, 21.6 to 39.6): 44.2% (panNET) and 16.4% (GI-NET). The median (range) duration of response was 19.9 (8.4-30.8) and 33.9 (10.6-38.3) months in the panNET and GI-NET groups, respectively. The median progression-free survival was 15.7 months (95% CI, 14.1 to 19.5). The most common adverse events were fatigue, hypertension, and diarrhea; 93.7% of patients required dose reductions or interruptions., Conclusion: We report the highest centrally confirmed response reported to date with a multikinase inhibitor in advanced GEP-NETs, with a particularly strong response in the panNET cohort. This study provides novel evidence for the efficacy of lenvatinib in patients with disease progression following treatment with other TAs, suggesting the potential value of lenvatinib in the treatment of advanced GEP-NETs., Competing Interests: Jaume CapdevilaConsulting or Advisory Role: Bayer, Eisai, Sanofi, Exelixis, Novartis, Ipsen, Pfizer, Merck Serono, Advanced Accelerator Applications, LillySpeakers' Bureau: Bayer, Eisai, Sanofi, Novartis, Ipsen, Pfizer, Merck Serono, LillyResearch Funding: Eisai, AstraZeneca, Advanced Accelerator Applications, Novartis, Ipsen, Bayer, PfizerTravel, Accommodations, Expenses: Pfizer, Ipsen, Eisai Nicola FazioHonoraria: Novartis, Ipsen, Merck, Sanofi, Advanced Accelerator ApplicationsConsulting or Advisory Role: Novartis/Ipsen, Advanced Accelerator Applications, Pfizer, Ipsen, Merck Serono, MSD OncologyResearch Funding: Novartis, Merck Serono, Ipsen, MSDOther Relationship: Springer, Il Pensiero Scientifico Editore Carlos Lopez LopezHonoraria: Roche, Merck, Sanofi, Novartis, Pfizer, Eisai, Ipsen, Bayer, AstraZeneca, Servier, Bristol Myers Squibb, MSD Oncology, Advanced Accelerator ApplicationsConsulting or Advisory Role: Amgen, Roche, Sanofi, Merck, Servier, Pfizer, Ipsen, Bayer, Eisai, AstraZenecaResearch Funding: Amgen, Roche, Merck, Merck Sharp & Dohme, AstraZeneca Spain, Sanofi, Bayer, Ipsen, Eisai, Bristol Myers Squibb, Boehringer IngelheimTravel, Accommodations, Expenses: Roche, Pfizer, Merck, Servier, Amgen, Ipsen Alex TeuleHonoraria: Ipsen, Novartis, AAA HealthCare, PfizerConsulting or Advisory Role: AAA HealthCare Juan ValleHonoraria: IpsenConsulting or Advisory Role: Ipsen, Novartis, AstraZeneca, Merck, Agios, Pfizer, PCI Biotech, Incyte, Keocyt, QED Therapeutics, Pieris Pharmaceuticals, Genoscience Pharma, Mundipharma EDO GmbH, Wren Laboratories, Nucana, Servier, Debiopharm Group, Imaging Equipment Limited, Hutchison MediPharma, Zymeworks, Aptitude Health, Sirtex Medical, BaxterSpeakers' Bureau: Novartis, Ipsen, Nucana, Imaging Equipment Limited, Mylan, IncyteTravel, Accommodations, Expenses: Nucana, Pfizer, Lilly Nicholas ReedHonoraria: Novartis, Ipsen, Eisai, Roche, LillyConsulting or Advisory Role: Novartis, Ipsen, EisaiSpeakers' Bureau: RocheResearch Funding: Novartis, Ipsen, Exelixis, Lilly Enrique GrandeHonoraria: Pfizer, Bristol Myers Squibb, Ipsen, Roche, Eisai, Eusa Pharma, MSD, Genzyme, Advanced Accelerator Applications, Novartis, Pierre Fabre, Lexicon, Celgene, Janssen-Cilag, Astellas Pharma, AstraZeneca, Lilly, EUSA PharmaConsulting or Advisory Role: MSD, Pfizer, Ipsen, Roche, Bristol Myers SquibbResearch Funding: Roche, Pfizer, AstraZeneca, Ipsen, Molecular Templates, Lexicon, Astellas PharmaTravel, Accommodations, Expenses: Bristol Myers Squibb, Roche/Genentech, Pfizer, Janssen-Cilag, Ipsen Rocio Garcia-CarboneroHonoraria: Ipsen, Roche, Sanofi, Servier, Novartis, Pfizer, Merck, PharmaMar, Advanced Accelerator Applications, Bristol Myers Squibb, AstraZeneca, Lilly, Boehringer Ingelheim, Gilead Sciences, Sysmex, Pierre Fabre, Midatech Pharma, Advanz Pharma, HMP, Bayer, MSDConsulting or Advisory Role: Ipsen, Novartis, Pfizer, HMP, Advanced Accelerator Applications, Bayer, PharmaMar, Merck, MSD, Pierre FabreResearch Funding: Pfizer, Bristol Myers Squibb, MSDTravel, Accommodations, Expenses: Roche, Merck Paula J FonsecaOther Relationship: Ipsen Jorge HernandoSpeakers' Bureau: Eisai, Ipsen, Roche, Acraf, Advanced Accelerator ApplicationsTravel, Accommodations, Expenses: Ipsen, Novartis, Advanced Accelerator Applications, Roche, AstraZeneca, Eisai Francesca SpadaConsulting or Advisory Role: Ipsen, Novartis, Advanced Accelerator ApplicationsSpeakers' Bureau: Ipsen, Novartis, Advanced Accelerator ApplicationsTravel, Accommodations, Expenses: Ipsen, Novartis, Advanced Accelerator Applications Vicente AlonsoConsulting or Advisory Role: Amgen, Sanofi/Regeneron, Ipsen, Servier, Merck SeronoTravel, Accommodations, Expenses: Merck Serono, Roche, Novartis, Amgen, Sanofi/Regeneron, Ipsen, Servier Alfredo BerrutiConsulting or Advisory Role: Janssen-Cilag, Astellas Pharma, AmgenSpeakers' Bureau: Janssen-Cilag, Astellas PharmaResearch Funding: Astellas Pharma, Janssen-CilagTravel, Accommodations, Expenses: Janssen-Cilag, Sanofi, Sanofi Adelaida La CastaTravel, Accommodations, Expenses: Amgen, Roche, MSD Oncology Sevilla IsabelConsulting or Advisory Role: Ipsen, Pfizer, AmgenSpeakers' Bureau: Ipsen, PharmaMar, Sanofi, AAA HealthCareTravel, Accommodations, Expenses: Ipsen Patrizia KumpResearch Funding: Bristol Myers SquibbTravel, Accommodations, Expenses: Ipsen Pablo GajateConsulting or Advisory Role: Ipsen, Roche, EisaiSpeakers' Bureau: IPSEN, Pfizer, Novartis, EisaiTravel, Accommodations, Expenses: Ipsen, Pfizer Ignacio MatosSpeakers' Bureau: MSDResearch Funding: ESMO Angela LamarcaConsulting or Advisory Role: EISAI, Nutricia, Ipsen, QED Therapeutics, RocheSpeakers' Bureau: Merck, Ipsen, Pfizer, Novartis, Incyte, Advanced Accelerator ApplicationsResearch Funding: Ipsen, RocheTravel, Accommodations, Expenses: Abbott Nutrition, Ipsen, Pfizer, Celgene, Novartis, Advanced Accelerator Applications, Sirtex Medical, Bayer, Delcath Systems, Mylan, NanoString Technologies Toni IbrahimConsulting or Advisory Role: EISAI, Novartis, SanofiTravel, Accommodations, Expenses: Ipsen, Pharmamar, NovartisNo other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
224. Adjuvant Nivolumab versus Placebo in Muscle-Invasive Urothelial Carcinoma.
- Author
-
Bajorin DF, Witjes JA, Gschwend JE, Schenker M, Valderrama BP, Tomita Y, Bamias A, Lebret T, Shariat SF, Park SH, Ye D, Agerbaek M, Enting D, McDermott R, Gajate P, Peer A, Milowsky MI, Nosov A, Neif Antonio J Jr, Tupikowski K, Toms L, Fischer BS, Qureshi A, Collette S, Unsal-Kacmaz K, Broughton E, Zardavas D, Koon HB, and Galsky MD
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents, Immunological adverse effects, B7-H1 Antigen metabolism, Carcinoma, Transitional Cell pathology, Carcinoma, Transitional Cell surgery, Chemotherapy, Adjuvant, Disease-Free Survival, Double-Blind Method, Female, Humans, Intention to Treat Analysis, Male, Middle Aged, Neoplasm Invasiveness, Neoplasm Staging, Nivolumab adverse effects, Placebos therapeutic use, Quality of Life, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms surgery, Antineoplastic Agents, Immunological therapeutic use, Carcinoma, Transitional Cell drug therapy, Nivolumab therapeutic use, Urinary Bladder Neoplasms drug therapy
- Abstract
Background: The role of adjuvant treatment in high-risk muscle-invasive urothelial carcinoma after radical surgery is not clear., Methods: In a phase 3, multicenter, double-blind, randomized, controlled trial, we assigned patients with muscle-invasive urothelial carcinoma who had undergone radical surgery to receive, in a 1:1 ratio, either nivolumab (240 mg intravenously) or placebo every 2 weeks for up to 1 year. Neoadjuvant cisplatin-based chemotherapy before trial entry was allowed. The primary end points were disease-free survival among all the patients (intention-to-treat population) and among patients with a tumor programmed death ligand 1 (PD-L1) expression level of 1% or more. Survival free from recurrence outside the urothelial tract was a secondary end point., Results: A total of 353 patients were assigned to receive nivolumab and 356 to receive placebo. The median disease-free survival in the intention-to-treat population was 20.8 months (95% confidence interval [CI], 16.5 to 27.6) with nivolumab and 10.8 months (95% CI, 8.3 to 13.9) with placebo. The percentage of patients who were alive and disease-free at 6 months was 74.9% with nivolumab and 60.3% with placebo (hazard ratio for disease recurrence or death, 0.70; 98.22% CI, 0.55 to 0.90; P<0.001). Among patients with a PD-L1 expression level of 1% or more, the percentage of patients was 74.5% and 55.7%, respectively (hazard ratio, 0.55; 98.72% CI, 0.35 to 0.85; P<0.001). The median survival free from recurrence outside the urothelial tract in the intention-to-treat population was 22.9 months (95% CI, 19.2 to 33.4) with nivolumab and 13.7 months (95% CI, 8.4 to 20.3) with placebo. The percentage of patients who were alive and free from recurrence outside the urothelial tract at 6 months was 77.0% with nivolumab and 62.7% with placebo (hazard ratio for recurrence outside the urothelial tract or death, 0.72; 95% CI, 0.59 to 0.89). Among patients with a PD-L1 expression level of 1% or more, the percentage of patients was 75.3% and 56.7%, respectively (hazard ratio, 0.55; 95% CI, 0.39 to 0.79). Treatment-related adverse events of grade 3 or higher occurred in 17.9% of the nivolumab group and 7.2% of the placebo group. Two treatment-related deaths due to pneumonitis were noted in the nivolumab group., Conclusions: In this trial involving patients with high-risk muscle-invasive urothelial carcinoma who had undergone radical surgery, disease-free survival was longer with adjuvant nivolumab than with placebo in the intention-to-treat population and among patients with a PD-L1 expression level of 1% or more. (Funded by Bristol Myers Squibb and Ono Pharmaceutical; CheckMate 274 ClinicalTrials.gov number, NCT02632409.)., (Copyright © 2021 Massachusetts Medical Society.)
- Published
- 2021
- Full Text
- View/download PDF
225. Case Report: Re-Treatment With Lu-DOTATATE in Neuroendocrine Tumors.
- Author
-
Vida Navas EM, Martínez Lorca A, Sancho Gutiérrez A, Sanz Gómez L, Navarro Martínez T, Grande Pulido E, Carrato Mena A, and Gajate Borau P
- Subjects
- Adult, Antineoplastic Agents therapeutic use, Bone Neoplasms diagnostic imaging, Bone Neoplasms drug therapy, Bone Neoplasms secondary, Everolimus therapeutic use, Humans, Magnetic Resonance Imaging, Male, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors secondary, Octreotide therapeutic use, Rectal Neoplasms diagnostic imaging, Rectal Neoplasms drug therapy, Rectal Neoplasms pathology, Retreatment, Sunitinib therapeutic use, Tomography, X-Ray Computed, Bone Neoplasms radiotherapy, Neuroendocrine Tumors radiotherapy, Octreotide analogs & derivatives, Organometallic Compounds therapeutic use, Radiopharmaceuticals therapeutic use, Rectal Neoplasms radiotherapy
- Abstract
Peptide receptor radionuclide therapy (PRRT) is an established treatment in advanced neuroendocrine tumors (NETs), which overexpressed somatostatin receptors. However, after progression there are a limited number of available treatments. We want to share a case report about a patient with a NET re-treated with
177 Lu-DOTATATE and a literature review about salvage treatment with PRRT. We present a 26-year-old man who started with pelvic pain and after a biopsy of a retro-rectal mass observed in a magnetic resonance was diagnosed with an advanced neuroendocrine tumour. After progression to lanreotide, everolimus and sunitinib, treatment with177 Lu-DOTATATE was initiated, achieving an excellent response with a progression free survival (PFS) of 38 months. At the time of progression, re-treatment with177 Lu-DOTATATE was decided, showing a new partial response, which is currently stable after 15 months. The patient had not presented significant treatment-related toxicity. Although there are no randomized phase III trials or a consensus about the number or dose of cycles, there is evidence about the efficacy and low toxicity of salvage treatment with177 Lu-DOTATATE in NETs. Median progression-free survival ranges from 6 to 22 months. Toxicity is mostly hematologic (anemia and neutropenia), 4-7% grade 3/4., Competing Interests: EG has served as advisor and delivered lectures for Pfizer, Bristol-Myers Squibb, Ipsen, Roche, Eisai, Eusa Pharma, MerckSharp&Dohme, Sanofi-Genzyme, Adacap, Novartis, PierreFabre, Lexicon and Celgene. PG has served as advisor and delivered lectures for Pfizer, Bristol-Myers Squibb, Ipsen, Roche, Eisai, Sanofi-Genzyme, Adacap, Novartis. The remaining 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., (Copyright © 2021 Vida Navas, Martínez Lorca, Sancho Gutiérrez, Sanz Gómez, Navarro Martínez, Grande Pulido, Carrato Mena and Gajate Borau.)- Published
- 2021
- Full Text
- View/download PDF
226. Spartalizumab in metastatic, well/poorly-differentiated neuroendocrine neoplasms.
- Author
-
Yao JC, Strosberg J, Fazio N, Pavel ME, Bergsland E, Ruszniewski P, Halperin DM, Li D, Tafuto S, Raj N, Campana D, Hijioka S, Raderer M, Guimbaud R, Gajate P, Pusceddu S, Reising A, Degtyarev E, Shilkrut M, Eddy S, and Singh S
- Abstract
Spartalizumab, a humanized anti-programmed death protein 1 (PD-1) monoclonal antibody, was evaluated in patients with well-differentiated metastatic grade 1/2 neuroendocrine tumors (NET) and poorly-differentiated gastroenteropancreatic neuroendocrine carcinomas (GEP-NEC). In this phase II, multicenter, single-arm study, patients received spartalizumab 400 mg every 4 weeks until confirmed disease progression or unacceptable toxicity. The primary endpoint was confirmed overall response rate (ORR) according to blinded independent review committee using response evaluation criteria in solid tumors 1.1. The study enrolled 95 patients in the NET group (30, 32 and 33 in the thoracic, gastrointestinal, and pancreatic cohorts, respectively), and 21 patients in the GEP-NEC group. The ORR was 7.4% (95% confidence interval [CI]: 3.0, 14.6) in the NET group (thoracic, 16.7%; gastrointestinal, 3.1%; pancreatic, 3.0%), which was below the predefined success criterion of ≥10%, and 4.8% (95% CI: 0.1, 23.8) in the GEP-NEC group. In the NET and GEP-NEC groups, the 12-month progression-free survival was 19.5% and 0%, respectively, and the 12-month overall survival was 73.5% and 19.1%, respectively. The ORR was higher in patients with ≥1% PD-L1 expression in immune/tumor cells or ≥1% CD8+ cells at baseline. The most common adverse events considered as spartalizumab-related included fatigue (29.5%) and nausea (10.5%) in the NET group, and increased aspartate and alanine aminotransferases (each 14.3%) in the GEP-NEC group. The efficacy of spartalizumab was limited in this heterogeneous and heavily pre-treated population; however, the results in the thoracic cohort is encouraging and warrants further investigation. Adverse events were manageable and consistent with previous experience.
- Published
- 2021
- Full Text
- View/download PDF
227. Practice change in the management of metastatic urothelial carcinoma after ASCO 2020.
- Author
-
Gajate P, Torres-Jiménez J, Bueno-Bravo C, and Couñago F
- Abstract
Metastatic urothelial carcinoma (mUC) is an incurable and aggressive disease. In the past decades there have been few effective treatment options that have impacted the prognosis of mUC patients. However, in the last few years, several drugs have emerged as new treatment choices that are changing the therapeutic landscape of mUC. Immune checkpoint inhibitors (ICIs) and targeted agents are useful treatment strategies that have been incorporated into our clinical practice. Nevertheless, cisplatin-based chemotherapy is still the standard of care in the first-line of metastatic disease. The results of the JAVELIN Bladder 100 phase 3 trial were presented at ASCO 2020, this trial evaluated the role of avelumab, an ICI, as maintenance therapy in patients who had not progressed after first-line platinum-based chemotherapy. The trial met its primary endpoint demonstrating an overall survival benefit with avelumab maintenance. In addition, new drugs and combinations are being evaluated to improve the outcomes of second and subsequent lines. Fibroblast growth factor receptor (FGFR) inhibitors and immunotherapy combinations were some of the strategies presented at ASCO 2020 that have shown promising results. Finally, the development of predictive biomarkers that help us in the decision-making process will be one of the most important challenges in the next years., Competing Interests: Conflict-of-interest statement: Gajate P has served as an advisor for Roche and Janssen, has served as a speaker for Pfizer, Roche and Janssen. Torres-Jimenez J, Bueno-Bravo C and Couñago F have nothing to disclose., (©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
228. Surgery of pancreatic metastasis from renal cell carcinoma.
- Author
-
Vilar Tabanera A, Muñoz Muñoz P, Molina Villar JM, Gajate P, and Sanjuanbenito A
- Published
- 2020
- Full Text
- View/download PDF
229. The PALBONET Trial: A Phase II Study of Palbociclib in Metastatic Grade 1 and 2 Pancreatic Neuroendocrine Tumors (GETNE-1407).
- Author
-
Grande E, Teulé A, Alonso-Gordoa T, Jiménez-Fonseca P, Benavent M, Capdevila J, Custodio A, Vera R, Munarriz J, La Casta A, Díez JJ, Gajate P, Molina-Cerrillo J, Matos I, Cristóbal EM, Ruffinelli JC, Palacios J, and García-Carbonero R
- Subjects
- Antineoplastic Combined Chemotherapy Protocols, Female, Humans, Male, Middle Aged, Piperazines adverse effects, Pyridines, Spain, Neuroendocrine Tumors drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Lessons Learned: Palbociclib demonstrated no detectable activity in molecularly unselected and heavily pretreated patients with advanced grade 1/2 pancreatic neuroendocrine tumors. Predictive biomarkers that improve patient selection should be investigated in future studies of palbociclib., Background: Palbociclib, a CDK4/6 inhibitor, has shown in vitro activity in pancreatic neuroendocrine tumor (pNET) cell lines. Here we prospectively assessed the activity and safety of palbociclib in monotherapy in metastatic refractory pNETs., Methods: This was a nonrandomized, open-label, phase II study of patients with metastatic grade (G)1/2 pNETs recruited from 10 centers in Spain. Palbociclib 125 mg was orally administered once daily for 21 of 28 days until disease progression or unacceptable toxicity., Results: Twenty-one patients were included; 52.4% were men, and median age was 57.4 years (range, 37.4-73.4). Patients had previously received a median of three prior lines of systemic therapy (range, 1-10) for advanced disease (somatostatin analogues, 71.4%; sunitinib, 81.0%; everolimus, 47.6%; chemotherapy, 47.6%). Nineteen patients were evaluated for objective response rate (ORR), with a median follow-up of 12.4 months (range, 7.53-19.33). No objective and confirmed responses were observed (0%); 11 (57.9%) patients had stable disease, and 6 of them lasted more than 6 months; 8 (42.1%) patients had disease progression as best response. Median progression-free survival (PFS) was 2.6 months (95% confidence interval [CI], 0-14.4) and median overall survival (OS) was 18.7 months (95% CI, 7.4-29.9; Fig. 1). Most frequent toxicities of any grade were asthenia (76.2%), neutropenia (42.9%), diarrhea (33.3%), and nausea (33.3%). Five (23.8%) patients developed G3-4 neutropenia and two (9.5%) patients developed G3-4 thrombocytopenia., Conclusion: Lack of activity was observed with palbociclib as a single agent in molecularly unselected and heavily pretreated patients with advanced G1/2 pNETs., (© AlphaMed Press; the data published online to support this summary are the property of the authors.)
- Published
- 2020
- Full Text
- View/download PDF
230. Rogaratinib in patients with advanced cancers selected by FGFR mRNA expression: a phase 1 dose-escalation and dose-expansion study.
- Author
-
Schuler M, Cho BC, Sayehli CM, Navarro A, Soo RA, Richly H, Cassier PA, Tai D, Penel N, Nogova L, Park SH, Schostak M, Gajate P, Cathomas R, Rajagopalan P, Grevel J, Bender S, Boix O, Nogai H, Ocker M, Ellinghaus P, and Joerger M
- Subjects
- Acute Kidney Injury chemically induced, Aged, Anorexia chemically induced, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Transitional Cell genetics, Diarrhea chemically induced, Fatigue chemically induced, Female, Humans, Hyperphosphatemia chemically induced, Hypoglycemia chemically induced, Lung Neoplasms genetics, Male, Maximum Tolerated Dose, Middle Aged, Piperazines adverse effects, Piperazines pharmacokinetics, Pyrroles adverse effects, Pyrroles pharmacokinetics, RNA, Messenger metabolism, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 1 genetics, Receptor, Fibroblast Growth Factor, Type 2 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 2 genetics, Receptor, Fibroblast Growth Factor, Type 3 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 3 genetics, Receptors, Fibroblast Growth Factor antagonists & inhibitors, Squamous Cell Carcinoma of Head and Neck genetics, Thiophenes adverse effects, Thiophenes pharmacokinetics, Vomiting chemically induced, Antineoplastic Agents administration & dosage, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Transitional Cell drug therapy, Lung Neoplasms drug therapy, Piperazines administration & dosage, Pyrroles administration & dosage, Receptors, Fibroblast Growth Factor genetics, Squamous Cell Carcinoma of Head and Neck drug therapy, Thiophenes administration & dosage
- Abstract
Background: The clinical activity of fibroblast growth factor receptor (FGFR) inhibitors seems restricted to cancers harbouring rare FGFR genetic aberrations. In preclinical studies, high tumour FGFR mRNA expression predicted response to rogaratinib, an oral pan-FGFR inhibitor. We aimed to assess the safety, maximum tolerated dose, recommended phase 2 dose, pharmacokinetics, and preliminary clinical activity of rogaratinib., Methods: We did a phase 1 dose-escalation and dose-expansion study of rogaratinib in adults with advanced cancers at 22 sites in Germany, Switzerland, South Korea, Singapore, Spain, and France. Eligible patients were aged 18 years or older, and were ineligible for standard therapy, with an Eastern Cooperative Oncology Group performance status of 0-2, a life expectancy of at least 3 months, and at least one measurable or evaluable lesion according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. During dose escalation, rogaratinib was administered orally twice daily at 50-800 mg in continuous 21-day cycles using a model-based dose-response analysis (continuous reassessment method). In the dose-expansion phase, all patients provided an archival formalin-fixed paraffin-embedded (FFPE) tumour biopsy or consented to a new biopsy at screening for the analysis of FGFR1-3 mRNA expression. In the dose-expansion phase, rogaratinib was given at the recommended dose for expansion to patients in four cohorts: urothelial carcinoma, head and neck squamous-cell cancer (HNSCC), non-small-cell lung cancer (NSCLC), and other solid tumour types. Primary endpoints were safety and tolerability, determination of maximum tolerated dose including dose-limiting toxicities and determination of recommended phase 2 dose, and pharmacokinetics of rogaratinib. Safety analyses were reported in all patients who received at least one dose of rogaratinib. Patients who completed cycle 1 or discontinued during cycle 1 due to an adverse event or dose-limiting toxicity were included in the evaluation of recommended phase 2 dose. Efficacy analyses were reported for all patients who received at least one dose of study drug and who had available post-baseline efficacy data. This ongoing study is registered with ClinicalTrials.gov, number NCT01976741, and is fully recruited., Findings: Between Dec 30, 2013, and July 5, 2017, 866 patients were screened for FGFR mRNA expression, of whom 126 patients were treated (23 FGFR mRNA-unselected patients in the dose-escalation phase and 103 patients with FGFR mRNA-overexpressing tumours [52 patients with urothelial carcinoma, eight patients with HNSCC, 20 patients with NSCLC, and 23 patients with other tumour types] in the dose-expansion phase). No dose-limiting toxicities were reported and the maximum tolerated dose was not reached; 800 mg twice daily was established as the recommended phase 2 dose and was selected for the dose-expansion phase. The most common adverse events of any grade were hyperphosphataemia (in 77 [61%] of 126 patients), diarrhoea (in 65 [52%]), and decreased appetite (in 48 [38%]); and the most common grade 3-4 adverse events were fatigue (in 11 [9%] of 126 patients) and asymptomatic increased lipase (in 10 [8%]). Serious treatment-related adverse events were reported in five patients (decreased appetite and diarrhoea in one patient with urothelial carcinoma, and acute kidney injury [NSCLC], hypoglycaemia [other solid tumours], retinopathy [urothelial carcinoma], and vomiting [urothelial carcinoma] in one patient each); no treatment-related deaths occurred. Median follow-up after cessation of treatment was 32 days (IQR 25-36 days). In the expansion cohorts, 15 (15%; 95% CI 8·6-23·5) out of 100 evaluable patients achieved an objective response, with responses recorded in all four expansion cohorts (12 in the urothelial carcinoma cohort and one in each of the other three cohorts), and in ten (67%) of 15 FGFR mRNA-overexpressing tumours without apparent FGFR genetic aberration., Interpretation: Rogaratinib was well tolerated and clinically active against several types of cancer. Selection by FGFR mRNA expression could be a useful additional biomarker to identify a broader patient population who could be eligible for FGFR inhibitor treatment., Funding: Bayer AG., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
231. LONG-TERM EFFECT OF 177 LU-DOTATATE ON SEVERE AND REFRACTORY HYPOGLYCEMIA ASSOCIATED WITH MALIGNANT INSULINOMA.
- Author
-
Iglesias P, Martínez A, Gajate P, Alonso T, Navarro T, and Díez JJ
- Abstract
Objective: Malignant insulinoma is an extremely uncommon tumor that is usually accompanied by severe hypoglycemia that is difficult to manage. At this time, the long-term effect of
177 Lu-DOTATATE (lutetium [Lu-177]-DOTA-Tyr3-octreotate) on this tumor is not well known., Methods: We report a case of severe, life-threatening, and refractory hypoglycemia associated with malignant insulinoma treated with177 Lu-DOTATATE., Results: A 51-year-old woman was referred because of severe, life-threatening, and refractory hypoglycemia due to malignant insulinoma. The patient had been treated unsuccessfully with chemotherapy, targeted therapies, and symptomatic therapy with diazoxide, steroids, and somatostatin analogues without success.177 Lu-DOTATATE adequately controlled her hypoglycemia after the other conventional treatments failed., Conclusion:177 Lu-DOTATATE was effective in providing rapid and long-term symptomatic control of the hypoglycemia and significantly improved the quality of life of the patient., Competing Interests: DISCLOSURE The authors have no multiplicity of interest to disclose., (Copyright © 2019 AACE.)- Published
- 2019
- Full Text
- View/download PDF
232. Trabectedin for reversing platinum resistance and resensitization to platinum in patients with recurrent ovarian cancer.
- Author
-
Casado A, Callata HR, Manzano A, Marquina G, Alonso T, Gajate P, Sotelo M, Cabezas S, Fernández C, and Díaz-Rubio E
- Subjects
- Aged, Aged, 80 and over, Clinical Trials as Topic, Disease-Free Survival, Doxorubicin administration & dosage, Doxorubicin adverse effects, Drug Resistance, Neoplasm drug effects, Female, Humans, Middle Aged, Neoplasm Recurrence, Local pathology, Ovarian Neoplasms pathology, Platinum adverse effects, Retrospective Studies, Trabectedin adverse effects, Neoplasm Recurrence, Local drug therapy, Ovarian Neoplasms drug therapy, Platinum administration & dosage, Trabectedin administration & dosage
- Abstract
Aims: We evaluated trabectedin in patients with platinum-resistant/refractory and partially platinum-sensitive recurrent ovarian cancer and the outcomes after reintroduction of platinum., Methods: Twenty-seven patients (platinum-resistant/refractory n = 24/PPS; n = 3) treated with trabectedin were retrospectively analyzed., Results: Trabectedin resulted in an objective response rate (ORR) of 18.2% with a 59.1% of disease control rate (ORR plus stable disease). The median progression-free and overall survival were 3.0 and 21.3 months, respectively. Subsequently, 17 patients were retreated with platinum and yield an ORR of 41.2% and DCR of 47.0%. The median progression-free and overall survival after platinum rechallenge were 5.0 and 14.7 months, respectively., Conclusion: Our results suggest that trabectedin may contribute to resensitize tumor cells to platinum rechallenge.
- Published
- 2019
- Full Text
- View/download PDF
233. Huge recurrent gastric neuroendocrine tumor: a second-line chemotherapeutic dilemma.
- Author
-
Ribeiro MJM, Alonso T, Gajate P, Molina J, Barquin A, Perna C, and Grande E
- Abstract
Chemotherapy is considered "state of the art" for the treatment of poorly differentiated neuroendocrine neoplasms. Unfortunately, there is no standard effective post-first-line treatment for relapsing high-grade gastroenteropancreatic neuroendocrine neoplasms. We report the case of a patient with a gastric neuroendocrine carcinoma stage IV, with massive gastrointestinal bleeding at diagnosis. After the first line of platin-based chemotherapy a major tumoral response was documented, but the patient relapsed after 4 months. A second line of chemotherapy treatment was given, with the FOLFOX regimen, and the patient has been free of progression for almost 2 years. There is no second-line standard treatment accepted for this type of carcinoma, but 5-fluorouracil combined with oxaliplatin showed interesting antitumor activity., Competing Interests: Conflict of interest: None
- Published
- 2018
- Full Text
- View/download PDF
234. Translating new data to the daily practice in second line treatment of renal cell carcinoma: The role of tumor growth rate.
- Author
-
Grande E, Martínez-Sáez O, Gajate-Borau P, and Alonso-Gordoa T
- Abstract
The therapeutic options for patients with metastatic renal cell carcinoma (mRCC) have completely changed during the last ten years. With the sequential use of targeted therapies, median overall survival has increased in daily practice and now it is not uncommon to see patients surviving kidney cancer for more than four to five years. Once treatment fails with the first line targeted therapy, head to head comparisons have shown that cabozantinib, nivolumab and the combination of lenvatinib plus everolimus are more effective than everolimus alone and that axitinib is more active than sorafenib. Unfortunately, it is very unlikely that we will ever have prospective data comparing the activity of axitinib, cabozantinib, lenvatinib or nivolumab. It is frustrating to observe the lack of biomarkers that we have in this field, thus there is no firm recommendation about the optimal sequence of treatment in the second line. In the absence of reliable biomarkers, there are several clinical endpoints that can help physicians to make decisions for an individual patient, such as the tumor burden, the expected response rate and the time to achieve the response to each agent, the prior response to the agent administered, the toxicity profile of the different compounds and patient preference. Here, we propose the introduction of the tumor-growth rate (TGR) during first-line treatment as a new tool to be used to select the second line strategy in mRCC. The rapidness of TGR before the onset of the treatment reflects the variability between patients in terms of tumor growth kinetics and it could be a surrogate marker of tumor aggressiveness that may guide treatment decisions., Competing Interests: Conflict-of-interest statement: Grande E has served as advisor and delivered lectures for Pfizer, IPSEN, and Eisai; Martínez-Sáez O, Gajate-Borau P and Alonso-Gordoa T declares no conflict of interest related to this publication.
- Published
- 2017
- Full Text
- View/download PDF
235. Targeting HIF-2 α in clear cell renal cell carcinoma: A promising therapeutic strategy.
- Author
-
Martínez-Sáez O, Gajate Borau P, Alonso-Gordoa T, Molina-Cerrillo J, and Grande E
- Subjects
- Carcinoma, Renal Cell blood supply, Carcinoma, Renal Cell pathology, Dimerization, Humans, Indans pharmacology, Indans therapeutic use, Kidney Neoplasms blood supply, Kidney Neoplasms pathology, Molecular Targeted Therapy, Sulfones pharmacology, Sulfones therapeutic use, Basic Helix-Loop-Helix Transcription Factors antagonists & inhibitors, Basic Helix-Loop-Helix Transcription Factors metabolism, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell metabolism, Kidney Neoplasms drug therapy, Kidney Neoplasms metabolism
- Abstract
The loss of the Von Hippel-Lindau tumor suppressor (VHL) is a key oncogenic event in the vast majority of patients with clear cell renal cell carcinoma (ccRCC). With the loss of the VHL protein (pVHL) function, the hypoxia inducible factor α (HIF-α) accumulates inside the tumor cell and dimerizes with HIF-β. The HIF-α/HIF-β complex transcriptionally activates hundreds of genes promoting the adaptation to hypoxia that is implicated in tumor development. There is growing evidence showing that HIF-2α subunit has a central role in ccRCC over HIF-1α. Thus, efforts have been made to specifically target this pathway. PT2385 and PT2399 are first-in-class, orally available, small molecule inhibitors of HIF-2 that selectively disrupt the heterodimerization of HIF-2α with HIF-1β. Preclinical and clinical data indicate that these new molecules are effective in blocking cancer cell growth, proliferation, and tumor angiogenesis characteristic in ccRCC. Treatment with HIF-2α specific antagonists, either alone or in combination with immunotherapy or other antiangiogenic agents have the potential to transform the therapeutic landscape in this tumor in the future. Herein, we summarize the molecular background behind the use of HIF-2α inhibitors in ccRCC and give an overview of the development of new agents in this setting., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
236. Capecitabine and temozolomide in grade 1/2 neuroendocrine tumors: a Spanish multicenter experience.
- Author
-
Crespo G, Jiménez-Fonseca P, Custodio A, López C, Carmona-Bayonas A, Alonso V, Navarro M, Aller J, Sevilla I, Grande E, Gajate P, Alonso-Gordoa T, Matos I, Capdevila J, Nieto B, and Barriuso J
- Subjects
- Aged, Capecitabine administration & dosage, Dacarbazine administration & dosage, Dacarbazine analogs & derivatives, Female, Humans, Male, Middle Aged, Neoplasm Grading, Neuroendocrine Tumors mortality, Proportional Hazards Models, Spain, Survival Analysis, Temozolomide, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neuroendocrine Tumors diagnosis, Neuroendocrine Tumors drug therapy
- Abstract
Background & Methods: Capecitabine and temozolomide chemotherapy was used in 65 patients with grade 1/2 neuroendocrine tumors (NETs). 46 patients (70.8%) had pancreatic NETs (pNETs)., Results: Response rate was 47.7%, with two complete responses (3.1%), 29 partial responses (44.6%) and 27 patients (41.5%) achieved stable disease. Median progression-free survival was 16.1 months (95% CI: 10.7-21.6) and overall survival was 38.3 months (95% CI: 24.6-51.9). Differences in progression-free survival and overall survival between pNETs and non-pNETs were not found. Nine (13.8%) patients experienced grade 3/4 toxicities, mainly thrombocytopenia (10.8%) and neutropenia (7.7%)., Conclusion: This is the largest reported series of NETs treated with capecitabine and temozolomide in daily practice and shows that this combination is a promising treatment option for both grade 1/2 pNETs and non-pNETs.
- Published
- 2017
- Full Text
- View/download PDF
237. Validation of the International Metastatic Renal-Cell Carcinoma Database Consortium (IMDC) prognostic model for first-line pazopanib in metastatic renal carcinoma: the Spanish Oncologic Genitourinary Group (SOGUG) SPAZO study.
- Author
-
Pérez-Valderrama B, Arranz Arija JA, Rodríguez Sánchez A, Pinto Marín A, Borrega García P, Castellano Gaunas DE, Rubio Romero G, Maximiano Alonso C, Villa Guzmán JC, Puertas Álvarez JL, Chirivella González I, Méndez Vidal MJ, Juan Fita MJ, León-Mateos L, Lázaro Quintela M, García Domínguez R, Jurado García JM, Vélez de Mendizábal E, Lambea Sorrosal JJ, García Carbonero I, González del Alba A, Suárez Rodríguez C, Jiménez Gallego P, Meana García JA, García Marrero RD, Gajate Borau P, Santander Lobera C, Molins Palau C, López Brea M, Fernández Parra EM, Reig Torras O, Basterretxea Badiola L, Vázquez Estévez S, and González Larriba JL
- Subjects
- Adult, Aged, Carcinoma, Renal Cell pathology, Databases, Factual, Disease-Free Survival, Female, Humans, Indazoles, Kaplan-Meier Estimate, Male, Middle Aged, Pyrimidines adverse effects, Retrospective Studies, Risk Factors, Spain, Sulfonamides adverse effects, Carcinoma, Renal Cell drug therapy, Molecular Targeted Therapy, Prognosis, Pyrimidines administration & dosage, Sulfonamides administration & dosage
- Abstract
Background: Patients with metastatic renal carcinoma (mRCC) treated with first-line pazopanib were not included in the International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) prognostic model. SPAZO (NCT02282579) was a nation-wide retrospective observational study designed to assess the effectiveness and validate the IMDC prognostic model in patients treated with first-line pazopanib in clinical practice., Patients and Methods: Data of 278 patients, treated with first-line pazopanib for mRCC in 34 centres in Spain, were locally recorded and externally validated. Mean age was 66 years, there were 68.3% male, 93.5% clear-cell type, 74.8% nephrectomized, and 81.3% had ECOG 0-1. Metastatic sites were: lung 70.9%, lymph node 43.9%, bone 26.3%, soft tissue/skin 20.1%, liver 15.1%, CNS 7.2%, adrenal gland 6.5%, pleura/peritoneum 5.8%, pancreas 5%, and kidney 2.2%. After median follow-up of 23 months, 76.4% had discontinued pazopanib (57.2% due to progression), 47.9% had received second-line targeted therapy, and 48.9% had died., Results: According to IMDC prognostic model, 19.4% had favourable risk (FR), 57.2% intermediate risk (IR), and 23.4% poor risk (PR). No unexpected toxicities were recorded. Response rate was 30.3% (FR: 44%, IR: 30% PR: 17.3%). Median progression-free survival (whole population) was 11 months (32 in FR, 11 in IR, 4 in PR). Median and 2-year overall survival (whole population) were 22 months and 48.1%, respectively (FR: not reached and 81.6%, IR: 22 and 48.7%, PR: 7 and 18.8%). These estimations and their 95% confidence intervals are fully consistent with the outcomes predicted by the IMDC prognostic model., Conclusion: Our results validate the IMDC model for first-line pazopanib in mRCC and confirm the effectiveness and safety of this treatment., (© The Author 2015. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
238. Influence of KRAS p.G13D mutation in patients with metastatic colorectal cancer treated with cetuximab.
- Author
-
Gajate P, Sastre J, Bando I, Alonso T, Cillero L, Sanz J, Caldés T, and Díaz-Rubio E
- Subjects
- Aged, Antibodies, Monoclonal, Humanized, Cetuximab, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Staging, Prognosis, Proto-Oncogene Proteins p21(ras), Retrospective Studies, Survival Rate, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Colorectal Neoplasms mortality, Mutation genetics, Proto-Oncogene Proteins genetics, ras Proteins genetics
- Abstract
Background: Patients with metastatic colorectal cancer (mCRC) with activating mutations at codon 12 or 13 of the KRAS gene are currently excluded from treatment with monoclonal antibodies against the epidermal growth factor receptor (EGFR), for example, cetuximab. Occasionally, some of these patients benefit from treatment with cetuximab, especially patients with a mutation at codon 13. We conducted an analysis to study the influence of the KRAS p.G13D mutation in patients with mCRC who were treated with cetuximab., Materials and Methods: We analyzed the KRAS mutation status of 110 patients who were treated with cetuximab between September 2003 and October 2008 at Hospital Clínico, San Carlos. We compared progression-free survival, overall survival, and response rate according to KRAS mutation status., Results: Patients with mutations at codon 13 compared with those with other KRAS mutations showed no statistically significant differences in progression-free survival (4.96 months [95% CI, 3.04-6.89 months] vs. 3.10 months [95% CI, 1.58-4.61 months]; hazard ratio [HR] 0.88 [95% CI, 44-1.75]; P = .72) and overall survival (8.2 months [95% CI, 4.2-12.1 months] vs. 14.6 months [95% CI, 8.0-21.2 months]; HR 0.50 [95% CI, 0.23-1.09]; P = .084). Patients with KRAS wild-type tumors have a longer progression-free survival (7.30 months [95% CI, 4.48-10.12 months]; HR 0.46 [95% CI, 0.23-0.91]; P = .025) and overall survival (19.0 months [95% CI, 10.2-27.8 months]; HR 0.32 [95% CI, 0.15-0.69]; P = .004) than patients with p.G13D-mutated tumors. Differences in the response rate were not observed between groups., Conclusion: Patients with mCRC and mutation at codon 13 of the KRAS gene do not appear to benefit from treatment with cetuximab. These results support the current clinical practice., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
239. Asthma related to Alternaria sensitization: an analysis of skin-test and serum-specific IgE efficiency based on the bronchial provocation test.
- Author
-
Fernández C, Bevilacqua E, Fernández N, Gajate P, de la Cámara AG, Garcimartín M, Vives R, and Rodríguez J
- Subjects
- Adolescent, Asthma microbiology, Female, Humans, Immunoglobulin E blood, Male, Predictive Value of Tests, ROC Curve, Regression Analysis, Retrospective Studies, Sensitivity and Specificity, Young Adult, Alternaria immunology, Asthma immunology, Bronchial Provocation Tests methods, Immunoglobulin E immunology, Skin Tests methods
- Abstract
Background: It is difficult to find a causal relationship between exposure to Alternaria spores and the development of asthma symptoms in sensitized individuals due to the complexity of clinical situations in which positive diagnostic tests are often found., Objective: To analyse the diagnostic efficiency of skin testing (ST) and serum-specific IgE to Alternaria, based on the results of a bronchial specific challenge with Alternaria extracts., Methods: Seventy-four asthmatic patients sensitized to Alternaria underwent a specific bronchial challenge with this mould. Skin-testing weal sizes, serum-specific IgE values (CAP-system) and bronchial challenge results were analysed by receiver operating characteristics curves (ROC curves) and logistic regression. The sensitivity, specificity, positive and negative predictive values were calculated for different cut-off points., Results: Bronchial challenges to Alternaria elicited a positive result in 45 patients (61%). Skin prick testing almost perfectly predicted the outcome of bronchoprovocation tests (area under the ROC curve of 0.957), whereas intradermal skin testing had moderate efficacy. A negative result for skin prick test (SPT) showed a 4% probability of a positive bronchial challenge in the logistic regression analysis. However, weals around 5.5 mm in diameter had 90% probability of a positive challenge. Quantification of serum-specific IgE correctly classified 86% of the cases. In the logistic regression analysis, a CAP value 16 kU(A)/L predicted a positive bronchial challenge result with 99% accuracy, whereas for a CAP value <0.35 kU(A)/L, this probability was 33%., Conclusions and Clinical Relevance: Most asthmatic patients with positive SPT results to Alternaria would have a positive bronchial challenge. As atmospheric mould levels may vary significantly with the weather conditions, sensitized patients should be instructed on the risk situations, environmental control measures and the importance of correct medication compliance. Immunotherapy with Alternaria could also be taken into account as a valid therapeutic option., (© 2010 Blackwell Publishing Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
240. Triatoma infestans in Greater Buenos Aires, Argentina.
- Author
-
Gajate P, Pietrokovsky S, Abramo Orrego L, Pérez O, Monte A, Belmonte J, and Wisnivesky-Colli C
- Subjects
- Animals, Argentina epidemiology, Chagas Disease prevention & control, Chagas Disease transmission, Chickens, Female, Humans, Male, Population Dynamics, Rabbits, Chagas Disease epidemiology, Housing, Animal, Insect Vectors growth & development, Triatoma growth & development
- Abstract
The Health Administration Agencies of many municipalities in Greater Buenos Aires (GBA) receive frequent reports on triatomines in houses. The aim of this work was to identify and describe the dispersal foci of Triatoma infestans in an urban neighborhood of GBA, and contribute to the knowledge of the epidemiological situation in the region. In June 1998, potentially infested places were entomologically evaluated. T. infestans was only detected in a hen building for egg production, which housed approximately 6,000 birds. A total of 2,930 insects were collected. Density was about 9 triatomines/m(2). The proportions of fifth instar nymphs and adults were significantly higher than those of the other stages (p<0.001). The number of triatomines collected largely exceeded the highest domestic infestation found in one house from rural endemic areas of Argentina. Though triatomines were negative for Trypanosoma cruzi, they could acquire the parasite by coming in contact with infected people living in GBA. Besides, the numerous and widely distributed places housing hens and chickens, would favor the settlement of the vector. Together, both facts may constitute a risk of parasitic vectorial transmission. It is recommended to intensify systematic activities of vector search and case detection in GBA.
- Published
- 2001
- Full Text
- View/download PDF
241. Comparative meiotic studies in Triatoma sordida (Stål) and T. guasayana Wygodzinsky & Abalos (Reduviidae, Heteroptera).
- Author
-
Rebagliati P, Papeschi AG, Mola LM, Pietrokovsky S, Gajate P, Bottazzi V, and Wisnivesky-Colli C
- Subjects
- Animals, Argentina, Chromosome Mapping, Karyotyping, Male, Meiosis genetics, Sex Chromosomes genetics, Spermatogenesis genetics, Triatoma classification, Triatoma cytology, Triatoma genetics, Triatominae classification, Triatominae cytology, Triatominae genetics
- Abstract
Triatoma sordida and T. guasayana are competent Trypanosoma cruzi vectors, with overlapping distribution areas in Argentina. Both species are morphologically similar, and their immature stages are hard to discriminate. Cytogenetic studies in the genus Triatoma reveal scarce karyotypic variations, being 2n = 20 + XY the most frequent diploid number in males. In the present work the meiotic behaviour of different Argentinian populations of T. sordida and T. guasayana has been analyzed; the meiotic karyotype of both species has also been compared. The species differ in total chromosome area and in the relative area of the sex chromosomes. These meiotic karyotypic differences constitute an additional tool for the taxonomic characterization of T. sordida and T. guasayana. The analysis of an interpopulation hybrid of T. sordida (Brazil x Argentina) reveals a regular meiotic behaviour; despite the presence of heteromorphic bivalents. Our observations support the hypothesis that karyotype variations through the gain or loss of heterochromatin can not be considered as a primary mechanism of reproductive isolation in Triatoma.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.