Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
NPJ Precis Oncol ; 8(1): 82, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561473

ABSTRACT

Pancreatic acinar cell carcinoma (PACC) is a rare form of pancreatic cancer that commonly harbors targetable alterations, including activating fusions in the MAPK pathway and loss-of-function (LOF) alterations in DNA damage response/homologous recombination DNA repair-related genes. Here, we describe a patient with PACC harboring both somatic biallelic LOF of NBN and an activating NTRK1 fusion. Upon disease progression following 13 months of treatment with folinic acid, fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX), genomic analysis of a metastatic liver biopsy revealed the emergence of a novel reversion mutation restoring the reading frame of NBN. To our knowledge, genomic reversion of NBN has not been previously reported as a resistance mechanism in any tumor type. The patient was treated with, but did not respond to, targeted treatment with a selective NTRK inhibitor. This case highlights the complex but highly actionable genomic landscape of PACC and underlines the value of genomic profiling of rare tumor types such as PACC.

2.
Cancer Discov ; : OF1-OF12, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38587856

ABSTRACT

Adagrasib, an irreversible, selective KRASG12C inhibitor, may be an effective treatment in KRASG12C-mutated colorectal cancer, particularly when combined with an anti-EGFR antibody. In this analysis of the KRYSTAL-1 trial, patients with previously treated KRASG12C-mutated unresectable or metastatic colorectal cancer received adagrasib (600 mg twice daily) plus cetuximab. The primary endpoint was objective response rate (ORR) by blinded independent central review. Ninety-four patients received adagrasib plus cetuximab. With a median follow-up of 11.9 months, ORR was 34.0%, disease control rate was 85.1%, and median duration of response was 5.8 months (95% confidence interval [CI], 4.2-7.6). Median progression-free survival was 6.9 months (95% CI, 5.7-7.4) and median overall survival was 15.9 months (95% CI, 11.8-18.8). Treatment-related adverse events (TRAEs) occurred in all patients; grade 3-4 in 27.7% and no grade 5. No TRAEs led to adagrasib discontinuation. Exploratory analyses suggest circulating tumor DNA may identify features of response and acquired resistance. SIGNIFICANCE: Adagrasib plus cetuximab demonstrates promising clinical activity and tolerable safety in heavily pretreated patients with unresectable or metastatic KRASG12C-mutated colorectal cancer. These data support a potential new standard of care and highlight the significance of testing and identification of KRASG12C mutations in patients with colorectal cancer.

3.
J Clin Oncol ; 41(25): 4097-4106, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37099736

ABSTRACT

PURPOSE: Adagrasib, a KRASG12C inhibitor, has demonstrated clinical activity in patients with KRASG12C-mutated non-small-cell lung cancer (NSCLC) and colorectal cancer (CRC). KRASG12C mutations occur rarely in other solid tumor types. We report evaluation of the clinical activity and safety of adagrasib in patients with other solid tumors harboring a KRASG12C mutation. METHODS: In this phase II cohort of the KRYSTAL-1 study (ClinicalTrials.gov identifier: NCT03785249; phase Ib cohort), we evaluated adagrasib (600 mg orally twice daily) in patients with KRASG12C-mutated advanced solid tumors (excluding NSCLC and CRC). The primary end point was objective response rate. Secondary end points included duration of response, progression-free survival (PFS), overall survival, and safety. RESULTS: As of October 1, 2022, 64 patients with KRASG12C-mutated solid tumors were enrolled and 63 patients treated (median follow-up, 16.8 months). The median number of prior lines of systemic therapy was 2. Among 57 patients with measurable disease at baseline, objective responses were observed in 20 (35.1%) patients (all partial responses), including 7/21 (33.3%) responses in pancreatic and 5/12 (41.7%) in biliary tract cancers. The median duration of response was 5.3 months (95% CI, 2.8 to 7.3) and median PFS was 7.4 months (95% CI, 5.3 to 8.6). Treatment-related adverse events (TRAEs) of any grade were observed in 96.8% of patients and grade 3-4 in 27.0%; there were no grade 5 TRAEs. TRAEs did not lead to treatment discontinuation in any patients. CONCLUSION: Adagrasib demonstrates encouraging clinical activity and is well tolerated in this rare cohort of pretreated patients with KRASG12C-mutated solid tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Mutation
4.
N Engl J Med ; 388(1): 44-54, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36546659

ABSTRACT

BACKGROUND: Adagrasib, an oral small-molecule inhibitor of mutant KRAS G12C protein, has shown clinical activity in pretreated patients with several tumor types, including colorectal cancer. Preclinical studies suggest that combining a KRAS G12C inhibitor with an epidermal growth factor receptor antibody could be an effective clinical strategy. METHODS: In this phase 1-2, open-label, nonrandomized clinical trial, we assigned heavily pretreated patients with metastatic colorectal cancer with mutant KRAS G12C to receive adagrasib monotherapy (600 mg orally twice daily) or adagrasib (at the same dose) in combination with intravenous cetuximab once a week (with an initial loading dose of 400 mg per square meter of body-surface area, followed by a dose of 250 mg per square meter) or every 2 weeks (with a dose of 500 mg per square meter). The primary end points were objective response (complete or partial response) and safety. RESULTS: As of June 16, 2022, a total of 44 patients had received adagrasib, and 32 had received combination therapy with adagrasib and cetuximab, with a median follow-up of 20.1 months and 17.5 months, respectively. In the monotherapy group (43 evaluable patients), a response was reported in 19% of the patients (95% confidence interval [CI], 8 to 33). The median response duration was 4.3 months (95% CI, 2.3 to 8.3), and the median progression-free survival was 5.6 months (95% CI, 4.1 to 8.3). In the combination-therapy group (28 evaluable patients), the response was 46% (95% CI, 28 to 66). The median response duration was 7.6 months (95% CI, 5.7 to not estimable), and the median progression-free survival was 6.9 months (95% CI, 5.4 to 8.1). The percentage of grade 3 or 4 treatment-related adverse events was 34% in the monotherapy group and 16% in the combination-therapy group. No grade 5 adverse events were observed. CONCLUSIONS: Adagrasib had antitumor activity in heavily pretreated patients with metastatic colorectal cancer with mutant KRAS G12C, both as oral monotherapy and in combination with cetuximab. The median response duration was more than 6 months in the combination-therapy group. Reversible adverse events were common in the two groups. (Funded by Mirati Therapeutics; KRYSTAL-1 ClinicalTrials.gov number, NCT03785249.).


Subject(s)
Antineoplastic Agents , Cetuximab , Colorectal Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/administration & dosage , Cetuximab/adverse effects , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/secondary , Piperazines/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use
5.
J Clin Oncol ; 39(6): 599-607, 2021 02 20.
Article in English | MEDLINE | ID: mdl-33125309

ABSTRACT

PURPOSE: Metastatic uveal melanoma has poor overall survival (OS) and no approved systemic therapy options. Studies of single-agent immunotherapy regimens have shown minimal benefit. There is the potential for improved responses with the use of combination immunotherapy. PATIENTS AND METHODS: We conducted a phase II study of nivolumab with ipilimumab in patients with metastatic uveal melanoma. Any number of prior treatments was permitted. Patients received nivolumab 1 mg/kg and ipilimumab 3 mg/kg for four cycles, followed by nivolumab maintenance therapy for up to 2 years. The primary outcome of the study was overall response rate (ORR) as determined by RECIST 1.1 criteria. Progression-free survival (PFS), OS, and adverse events were also assessed. RESULTS: Thirty-five patients were enrolled, and 33 patients were evaluable for efficacy. The ORR was 18%, including one confirmed complete response and five confirmed partial responses. The median PFS was 5.5 months (95% CI, 3.4 to 9.5 months), and the median OS was 19.1 months (95% CI, 9.6 months to NR). Forty percent of patients experienced a grade 3-4 treatment-related adverse event. CONCLUSION: The combination regimen of nivolumab plus ipilimumab demonstrates activity in metastatic uveal melanoma, with deep and sustained confirmed responses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ipilimumab/therapeutic use , Melanoma/drug therapy , Nivolumab/therapeutic use , Uveal Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Female , Humans , Ipilimumab/pharmacology , Male , Middle Aged , Nivolumab/pharmacology
6.
Curr Treat Options Oncol ; 21(2): 10, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32025932

ABSTRACT

OPINION STATEMENT: Patients with clinical stage III melanoma, defined as palpable lymph nodes with or without in-transit metastases, have poor prognosis even with recent advances with targeted and checkpoint inhibitor therapy in the adjuvant setting. Neoadjuvant therapy for clinical stage III melanoma is an attractive treatment paradigm as patient outcomes may be improved by earlier introduction to systemic therapy. Additionally, preoperative therapy that shrinks disease has the potential to improve surgical morbidity. Neoadjuvant therapy also provides for pathologic response assessment which can serve as a way to stratify patient outcomes and subsequent disease relapse risk. Early trials of neoadjuvant immunotherapy are yielding promising results, with high rates of pathologic complete response (pCR) and improved relapse-free survival rates. Ipilimumab, nivolumab with or without ipilimumab, and pembrolizumab have been investigated in the neoadjuvant setting. A meta-analysis has shown a 1-year relapse-free survival rate of over 80% with neoadjuvant immunotherapy. Importantly, pooled data also shows that pCR strongly correlates with outcomes. Early phase trials have also highlighted the importance of dosing of neoadjuvant therapy to appropriately balance response and immune related toxicities, which can be severe. The combination of ipilimumab 1 mg/kg and nivolumab 3 mg/kg has been identified as an optimal regimen for further study. Translational studies have highlighted the ability of neoadjuvant immunotherapy to expand tumor-specific T cells in both the tumor microenvironment and peripheral blood. At this time, surgical resection and adjuvant therapy remains standard of care for clinical stage III melanoma; however, appropriate patients should be considered for ongoing neoadjuvant clinical trials.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy , Melanoma/pathology , Melanoma/therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Chemotherapy, Adjuvant , Clinical Trials as Topic , Combined Modality Therapy , Disease Management , Disease Susceptibility , Drug Evaluation, Preclinical , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/metabolism , Immunotherapy/adverse effects , Immunotherapy/methods , Melanoma/etiology , Melanoma/mortality , Neoadjuvant Therapy , Neoplasm Staging , Treatment Outcome
7.
Laryngoscope ; 130(4): E140-E143, 2020 04.
Article in English | MEDLINE | ID: mdl-31074845

ABSTRACT

Pembrolizumab, an anti-PD-1 checkpoint inhibitor, is used in patients with recurrent or metastatic squamous cell carcinoma of the head and neck who have received prior therapy with a platinum-based regimen. As a monotherapy, it is generally well tolerated, but a small percentage of patients may develop immune-mediated inflammatory reactions. We report a case of mucositis and laryngeal edema in a patient on pembrolizumab and review the literature. Laryngoscope, 130:E140-E143, 2020.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Hypopharyngeal Neoplasms/drug therapy , Mucositis/chemically induced , Squamous Cell Carcinoma of Head and Neck/drug therapy , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Humans , Male , Mucositis/drug therapy , Neoplasm Recurrence, Local , Steroids/therapeutic use
8.
Ther Adv Med Oncol ; 11: 1758835919830826, 2019.
Article in English | MEDLINE | ID: mdl-30815041

ABSTRACT

The development of BRAF and MEK inhibitors (BRAFis and MEKis) and immune checkpoint inhibitors have changed the management of advanced stage melanoma and improved the outcomes of patients with this malignancy. However, both therapeutic approaches have limitations, including a limited duration of benefit in subsets of BRAF-mutant melanoma patients treated with targeted therapy and a lower overall response rate without a clear predictive biomarker in patients treated with checkpoint inhibitors. Preclinical and translational data have shown that BRAFis and MEKis alter the tumor microenvironment to make it more amenable to immunotherapy and have provided the scientific rationale for combing BRAFis and MEKis with immunotherapy. In this review, the initial studies demonstrating the impact of BRAFis and MEKis on the expression of melanoma differentiation antigens, T-cell infiltration, and the balance of immune stimulatory and immune suppressive cells and cytokines are addressed. Preclinical work on the combination of targeted therapy with BRAFis and MEKis with immunotherapy are reviewed, highlighting improved tumor responses in mouse models of BRAF-mutated melanoma treated with combinatorial strategies. Lastly, data from early clinical trials of combined targeted therapy and immunotherapy are discussed, focusing on response rates and toxicities.

SELECTION OF CITATIONS
SEARCH DETAIL
...