GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 17 ( 2019-09-01), p. 5376-5387
    Abstract: Cancer treatment is limited by inaccurate predictors of patient-specific therapeutic response. Therefore, some patients are exposed to unnecessary side effects and delays in starting effective therapy. A clinical tool that predicts treatment sensitivity for individual patients is needed. Experimental Design: Patient-derived cancer organoids were derived across multiple histologies. The histologic characteristics, mutation profile, clonal structure, and response to chemotherapy and radiation were assessed using bright-field and optical metabolic imaging on spheroid and single-cell levels, respectively. Results: We demonstrate that patient-derived cancer organoids represent the cancers from which they were derived, including key histologic and molecular features. These cultures were generated from numerous cancers, various biopsy sample types, and in different clinical settings. Next-generation sequencing reveals the presence of subclonal populations within the organoid cultures. These cultures allow for the detection of clonal heterogeneity with a greater sensitivity than bulk tumor sequencing. Optical metabolic imaging of these organoids provides cell-level quantification of treatment response and tumor heterogeneity allowing for resolution of therapeutic differences between patient samples. Using this technology, we prospectively predict treatment response for a patient with metastatic colorectal cancer. Conclusions: These studies add to the literature demonstrating feasibility to grow clinical patient-derived organotypic cultures for treatment effectiveness testing. Together, these culture methods and response assessment techniques hold great promise to predict treatment sensitivity for patients with cancer undergoing chemotherapy and/or radiation.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1128-1128
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1128-1128
    Abstract: Background: The identification of treatment strategies targeting PIK3CA mutant colorectal cancer (CRC) are of great clinical interest. Previous work from our lab has identified MTORC1/2 and HDAC1/2 inhibition with copanlisib (cop; PI3K/MTOR) and romidepsin (romi; HDAC1/2) as a potential therapeutic strategy. We hypothesized that changes in c-MYC protein levels and c-MYC target gene (CTG) alterations might be a potential mechanism of action for this combination. Methods: Known CTGs were identified and their expression levels were examined in PIK3CA mutant vs WT CRCs using the cBioPortal Colorectal Adenocarcinoma (TCGA, PanCancer Atlas) dataset. Murine derived cancer organoids (MDCO) were generated from adenocarcinomas of Apc and Pik3ca mutant transgenic mice (F1 (FVB x B6) Apcfl/+ Pik3caH1047R/+). MDCO results were corroborated using the human 2D isogenic cell lines SW48 and SW48PIK3CA-H1047R (SW48PK) and RAS/RAF WT patient derived cancer organoids (PDCO) generated from CRC patient samples under approved IRB protocols. Immunoblots (IB) were used to assess c-MYC levels after treatment with cop, romi, and the combination across all models. RNA sequencing was conducted on PDCOs and SW48PK cells and changes in 16 CTGs were examined. An aggregate score was created for each treatment group where a statistically significantly altered CTG with log fold change ≥1.5 added one point and ≤-1.5 subtracted one point. All others were scored 0. Results: c-MYC target genes were assessed for differential expression in PIK3CA mutant CRC vs PIK3CA WT CRC with only 1/16 genes decreased in PIK3CA mutant CRC (GADD45A: log ratio -0.21, q=0.01). Next, MDCOs treated with the combination showed a decrease in total c-MYC levels in the combination therapy. These results were corroborated in two human 2D CRC cell lines, SW48 and SW48PK and a panel of PDCOs. Interestingly, c-MYC levels decreased in both romi alone and the combination in both the SW48 and SW48PK cell lines after 24 hours of treatment. However, the extent to which c-MYC levels were decreased was not as substantial in the panel of PDCOs. Across all in vitro models a decrease in PI3K signaling, as illustrated by decreased pRPS6 and p4EBP1 in response to cop treatment and an increase in H3K27 acetylation in response to romi, was observed in the single agent and combination therapies. RNA sequencing demonstrated that cop, romi, and combo had a CTG score of 0, -4, and -7 respectively in the PIK3CA mutant PDCO. Similar results were seen in an additional RAS/RAF WT PDCO (0, 1, and -4, respectively) and SW48PK cells (0, -3, and -7, respectively). Conclusion: A potential mechanism by which cop and romi treatment promote tumor response is through a decrease in c-MYC protein levels and expression of downstream c-MYC target genes. This regimen deserves further mechanistic investigations in vivo. Citation Format: Rebecca A. DeStefanis, Autumn M. Olson, Alyssa K. DeZeeuw, Susan N. Payne, Cheri A. Pasch, Linda Clipson, Dustin A. Deming. MTORC1/2 and HDAC1/2 inhibition promote tumor response through inhibition of MYC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1128.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2022-03-25)
    Abstract: Representative models are needed to screen new therapies for patients with cancer. Cancer organoids are a leap forward as a culture model that faithfully represents the disease. Mouse-derived cancer organoids (MDCOs) are becoming increasingly popular, however there has yet to be a standardized method to assess therapeutic response and identify subpopulation heterogeneity. There are multiple factors unique to organoid culture that could affect how therapeutic response and MDCO heterogeneity are assessed. Here we describe an analysis of nearly 3500 individual MDCOs where individual organoid morphologic tracking was performed. Change in MDCO diameter was assessed in the presence of control media or targeted therapies. Individual organoid tracking was identified to be more sensitive to treatment response than well-level assessment. The impact of different generations of mice of the same genotype, different regions of the colon, and organoid specific characteristics including baseline size, passage number, plating density, and location within the matrix were examined. Only the starting size of the MDCO altered the subsequent growth. These results were corroborated using ~ 1700 patient-derived cancer organoids (PDCOs) isolated from 19 patients. Here we establish organoid culture parameters for individual organoid morphologic tracking to determine therapeutic response and growth/response heterogeneity for translational studies.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2615211-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3048-3048
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3048-3048
    Abstract: Introduction: Colorectal cancer (CRC) treatment is becoming increasingly individualized as clinical mutation profiling is a standard of care. Oncogenic mutations of the PIK3CA gene induce a constitutively active phosphatidylinositol 3-kinase with downstream effects that promote tumor initiation and progression. Effective clinical therapeutic strategies targeting this subtype of colorectal cancer have remained elusive. Here we develop a high-throughput organoid drug screening platform to examine combination strategies targeting PIK3CA mutant CRC. Methods: Murine-derived CRC organoids (MDCOs) were derived from a transgenic mouse model of PIK3CA and APC mutated colorectal cancer (F1 (FVBxB6) Apcfl/+ Pik3caH1047R), matured for 48 hours and plated in 96 well plates. A total of 96 compounds chosen from clinically available anticancer drugs and select drugs in clinical development were analyzed for their efficacy in reducing organoid growth on their own, as well as in combination with copanlisib (PI3K/MTOR inhibitor) over 48 hours. Select treatments were confirmed using our standard 24 well plate assay. Additionally, irinotecan (SN38; topoisomerase-I inhibitor), panitumumab (EGFR inhibitor), and olaparib (PARP inhibitor) were investigated alone and in combination with copanlisib based on clinical use for this population or recent promising clinical trial data. Results: SN38, olaparib, and panitumumab yielded insignificant changes in spheroid size in the combination regimen compared to copanlisib alone (median relative change in organoid diameter - copanlisib -8%, p & lt;0.001; SN38 combo -8%, p=0.5; olaparib combo -13%, p=0.6; and panitumumab combo 1%, p=0.9). The MDCOs were successfully utilized for the high-throughput drug screen. Differential sensitivity was identified using change in organoid diameter without the need for reagents or dyes which could complicate the screening method. Of the 96 compounds investigated in the high-throughput screen, 12 resulted in enhanced treatment effect when combined with copanlisib. Three of these 12 compounds inhibit microtubule formation, and others inhibit components of the RAS pathway. Two of these drug combinations were examined further and significant treatment responses were confirmed. Conclusion: Advances in the understanding of the molecular basis of cancer allow for a more precise approach to cancer treatment. High-throughput techniques are needed to better identify novel treatment strategies for molecular subtypes. Here we describe a label free murine-derived cancer organoid method to screen drugs for combination treatment strategies. Citation Format: Alyssa K. DeZeeuw, Rebecca A. DeStefanis, Gioia Sha, Cheri A. Pasch, Linda Clipson, Dustin A. Deming. Murine-derived colorectal cancer organoid culture high-throughput drug screening to identify novel combinations targeting PIK3CA mutant cancers [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3048.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 2673-2673
    Abstract: Background: Treating colorectal cancer (CRC) using targeted therapies based on each patient’s individual mutational profile continues to gain interest. Oncogenic mutations of the PIK3CA gene leads to tumor initiation and progression. Copanlisib inhibits the PI3K pathway and has proven to be an effective treatment strategy for various cancers characterized by overactivation of the PI3K signaling cascade. However, resistance mechanisms to targeted therapies such as copanlisib are frequently encountered, and can be overcome through combination with another molecule such as romidepsin, a histone deacetylase (HDAC) inhibitor. Here we aim to identify a novel therapeutic regimen for improved treatment of the molecular subtypes of CRC that can be targeted with PI3K inhibition. Methods: Organoids were derived from colorectal cancer tumor tissue from two different biopsy sites from one patient with PIK3CA mutant rectal cancer (RC46A and RC46B), as well as from a patient with APC and TP53 mutant CRC (MTB74) under approved IRB protocols. All organoids were plated and allowed to mature for 24 to 48 hours before treatment and baseline images were obtained. The organoids were imaged again 48 hours later, and response was determined by measuring the change in organoid diameter using ImageJ. Additionally, organoids were collected for immunoblotting to examine alterations in the PI3K pathway as well as histone acetylation, and proteins involved in apoptosis. Results: Control organoids demonstrated increased median relative changes in diameter of +53.38%, +43.04% and +46.2% for RC46A, RC46B and MTB74, respectively. In comparison, the combination treatment group exhibited median relative changes for RC46A, RC46B and MTB74 as -29.19%, -46.46% and -100% (p & lt;0.001). These results were supported with immunoblot analysis that revealed a synergistic increase in histone acetylation with the combination treatment. Single agent copanlisib treatment elicited alterations in PI3K signaling including downregulation of pS6, pAKT, p4EBP1 as expected. Lastly, an increase in cleaved PARP was observed in combination treated organoids which is indicative of increased apoptosis. Conclusions: As molecular diagnostic technology continues to advance, treating each individual cancer with a targeted therapy based on molecular profiling is becoming more feasible. Dual inhibition of the PI3K pathway and HDAC is effective in reducing cell growth and proliferation of CRC organoids characterized by PIK3CA mutations, as well as other molecular subtypes such as cancers with APC and TP53 mutations. This combination strategy shows promise for improving treatment response and reducing resistance in patients across these molecular subtypes. Citation Format: Alyssa K. DeZeeuw, Rebecca A. DeStefanis, Susan N. Payne, Autumn M. Olson, Cheri A. Pasch, Linda Clipson, Dustin A. Deming. Combined inhibition of PI3K and HDAC1/2 as a novel treatment strategy for RAS/RAF wildtype colorectal cancer in a patient derived organoid model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2673.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3721-3721
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3721-3721
    Abstract: Introduction: Colorectal cancer (CRC) is a leading cause of cancer-related deaths. PIK3CA mutations are found in 18% of CRCs which lead to the constitutive activation of the PI3K/MTOR pathway and can promote tumorigenesis. Here, we examine the efficacy of the FDA approved inhibitors copanlisib, a PI3K/MTOR inhibitor, and romidepsin, a HDAC1/2 inhibitor, in CRC with a PIK3CA mutation. Methods: CRC mouse derived cancer organoids (MDCOs) were derived from Apc and Pik3ca mutant mice (Fc1Apcfl/+Pik3caH1047R/+). Brightfield images of the MDCOs were taken prior to treatment with copanlisib, romidepsin, or the combination and 48 hours post-treatment. The change in diameter of each organoid over the 48-hour treatment was determined. Additionally, immunoblotting was performed to confirm known targets of copanlisib and romidepsin were altered in response to drug treatment. MTORC1/2 and HDAC1/2 inhibition were also investigated in vivo. SW48 and SW48PIK3CA-H1047R (SW48PK) xenograft mice were treated with a vehicle control, copanlisib, romidepsin, or the combination therapy. Results: In the diameter analysis study, the combination therapy had the largest effect size compared to control (Glass’s Δ 2.82). Single agents copanlisib and romidepsin had smaller effect sizes (Glass’s Δ 1.75 and 2.04, respectively). Immunoblotting results indicated a decrease in phosphoAKT (Ser473) and pRPS6 (Ser235/236) in the copanlisib treated samples and an increase in H3K27 acetylation was seen in the romidepsin treated samples. There was also increased cleaved PARP, an indicator of apoptosis, in the romidepsin and combination therapy treatment groups. In vivo, SW48 xenografts showed a greater response in the combination therapy compared to either single agent therapy (median relative change in tumor volume: control=339%; combination therapy=107%(p-value & lt;0.05), copanlisib=225%(p-value=0.17), romidepsin=200%). A similar trend was seen in the SW48PK xenograft mice (median relative change in tumor volume: control=241%; combination therapy=70% (p-value & lt;0.05), copanlisib=132%(p-value & lt;0.05), romidepsin=177%). Conclusion: MDCOs with Apc and Pik3ca mutations had an increased response to treatment with the combination therapy as compared to the single agents alone. Additionally, in vivo studies with human CRC xenografts showed enhanced inhibition of tumor growth with both MTORC1/2 and HDAC1/2 inhibition. These data demonstrate potential for this combination treatment strategy for the treatment of PIK3CA mutant CRC and this combination warrants further investigation in other models and clinically. Citation Format: Autumn M. Olson, Rebecca A. DeStefanis, Alyssa K. DeZeeuw, Susan N. Payne, Cheri A. Pasch, Linda Clipson, Dustin A. Deming. MTORC1/2 and HDAC1/2 inhibition as therapy for colorectal cancer with PIK3CA mutation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3721.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. e15543-e15543
    Abstract: e15543 Background: Detection of circulating tumor DNA (ctDNA) following operative management for colorectal cancer (CRC) as part of molecular residual disease (MRD) assays has shown significant prognostic implications. The majority of studies examining the time to detection of cancer recurrence have been done retrospectively without the treating physician knowing the results of the assay. Limited prospective data exists regarding the probability of detecting cancer on imaging at the time of a positive MRD test following operative management of all sites of disease. Here, we analyze the association of positive MRD testing with imaging findings of recurrent or metastatic CRC using a cohort of patients that underwent prospective MRD testing. Methods: Patients with stage I-IV intestinal cancers at University of Wisconsin Carbone Cancer Center who underwent prospective Natera Signatera MRD testing following operative management of their disease were identified and consented to an IRB-approved registry protocol. Those consented patients who had a positive test without being on systemic therapy and had clinical imaging within 45 days were analyzed further. The presence of definitive cancer, indeterminate findings, and no evidence of cancer was recorded per the treating physician using standard of care clinical imaging. The presence of cancer on imaging was then correlated with the quantity of ctDNA (mean tumor molecules (MTM)/ml). Results: 163 patients with stage I-IV CRC and 1 small intestinal cancer underwent MRD testing. 30 positive ctDNA tests with imaging were available across 22 unique patients (median age 54 years (29-84) 7 females). 23 samples were in the resected metastatic setting, 5 stage III and 2 stage I. Cancer was seen on imaging in 19/30 (63%) of positive tests, indeterminate in 3 (10%) and negative for cancer in 9/30 (27%). With ctDNA levels 〉 10 MTM/ml (n = 7), cancer was detected on imaging in all cases. With ctDNA levels of 1-10 MTM/ml cancer was detected in 42% and at 〈 1 MTM/ml in 64%. A trend towards greater detection of cancer was observed when MRI or PET/CT was performed. Radiologic disease detection in patients with positive MRD analysis was higher in those with stage IV disease (70%) vs. stage I-III (43%). Conclusions: ctDNA MRD positivity is associated with high rates of detection of recurrent/metastatic CRC, especially in patients with resected stage IV disease and with a quantity of 〉 10 MTM/ml. Clinical imaging should be considered at the time of detection of ctDNA.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 75-75
    Abstract: Colorectal cancer (CRC) is a leading cause of cancer related death with PIK3CA mutations occurring in ~18% of all cases. Mutations in this gene lead to constitutive activation of the phosphoinositide-3 kinase (PI3K) oncogene. Previously we've shown that MTORC1/2 inhibition is sufficient to induce a therapeutic response both in vitro and in vivo with minimal induction of apoptosis. BCL-xL is a well-known negative regulator of apoptosis in solid tumors. We therefore investigated whether inhibition of the BCL-2 family, and more specifically BCL-xL, would enhance therapeutic response and induction of apoptosis. Murine-derived cancer organoids (MDCOs) were generated from invasive colon adenocarcinomas of Apc and Pik3ca transgenic mice (F1 (FVBxB6) Apcfl/+ Pik3caH1047R). MDCOs were allowed to mature for 24 hours, baseline brightfield imaging performed and therapeutic agents added at concentrations outlined below. Median relative change in organoid diameter after 48 hours of treatment was determined. In vivo response was measured in F1 (FVBxB6) Apcfl/+ Pik3caP110* mice as change in endoscopic tumor lumen occlusion over 14 days. Immunoblotting (IB) and immunofluorescence (IF) were utilized to evaluate for induction of apoptosis. Navitoclax (BCL-2/BCL-xL/BCL-w inhibitor, 250nM) was evaluated alone and in combination with a panel of MTORC1/2 inhibitors (BEZ-235 (BEZ), TAK-228 (TAK), copanlisib (Cop), 200nM). Navitoclax did not induce a treatment response as a single agent. Enhanced response was seen with the combination compared to the MTORC1/2 inhibitors alone (Bez 56% vs combo -100%, p & lt;0.001; TAK -27% vs combo -100%, p & lt;0.001; Cop -16% vs -100%; p & lt;0.001). Results were confirmed in vivo with BEZ-235 (30mg/kg/day), navitoclax (80mg/kg/day), or the combination with the greatest reduction in lumen occlusion of colon tumors in the combination therapy (control +15%, navitoclax +1%, BEZ -15%, and combo -42%, p & lt;0.003 BEZ vs combo). IB of cleaved PARP, a main cleavage target of cleaved caspase 3 (CC3) once apoptosis is induced, and IF of CC3 confirmed induction of apoptosis was highest in the combination therapy in both in vitro and in vivo studies. This induction was found as early as 6 hours post treatment in the MDCOs. To confirm inhibition of BCL-xL was the primary anti-apoptotic protein necessary for this induction of apoptosis, MDCOs were treated with copanlisib (200nM) alone or in combination with WEHI-539 (BCL-xL inhibitor, 250nM) or ABT-199 (BCL-2 inhibitor, 250nM). An enhanced sensitivity was observed when MTORC1/2 inhibition was combined with the inhibition of BCL-xL compared to BCL-2. These studies indicate that BCL-xL signaling reduces MTORC1/2 inhibitor response and targeting BCL-xL in combination with MTORC1/2 enhances both the treatment response and the induction of apoptosis in PIK3CA mutant CRC. Citation Format: Rebecca A. DeStefanis, Alyssa DeZeeuw, Gioia Sha, Susan N. Payne, Christopher P. Babiarz, Devon Miller, Demetra K. Korkos, Cheri A. Pasch, Linda Clipson, Kristina Matkowskyj, Dustin A. Deming. BCL-xL inhibition enhances therapeutic response of MTORC1/2 inhibition and induction of apoptosis in PIK3CA mutant colorectal cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 75.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. e15500-e15500
    Abstract: e15500 Background: Anal cancer (AC) is a relatively uncommon malignancy in the United States, however, this disease continues to increase in incidence, especially in individuals over 50 years old. Current standard of care for locoregional disease includes mitomycin and 5-fluorouracil (M/5FU) with radiation. This combination is associated with significant toxicity limiting its use in those patients who are elderly or have multiple comorbidities. Carboplatin and paclitaxel (C/P) is now the preferred standard of care treatment regimen for metastatic AC. Weekly C/P has become a standard of care option in combination with radiation for locally advanced esophageal cancer. Here we evaluate the efficacy of weekly C/P chemoradiation in treating a cohort of patients unable to receive standard M/5FU chemoradiation. Methods: Subjects were consented to an IRB approved AC registry at the University of Wisconsin Carbone Cancer Center. Patients (pts) were identified to have received weekly infusional C/P concurrent with standard-dose radiation for localized AC between January 1, 2016 and December 1, 2022. Clinical response was determined based on evidence of disease on imaging or anoscopy and toxicities were graded according to CTCAE v5. Results: 8 pts (7F/1M; median age 76 (54-87)) were found to meet eligibility criteria. 6 had perianal cancers and 2 had cancers within the anal canal. 6 had T2 disease and 2 had T3 tumors. 2 had node positive disease. Dosing ranged from C (AUC 1.5-2), P (30-50mg/m2) and radiation (50.4-58 Gy). Patients completed a median of 82.5% (40%-100%) of intended treatments. Most common toxicities included leukopenia, anemia, perianal pain, and fatigue. Grade 3 or higher toxicities included neutropenia (3) of which 2 resulted in febrile neutropenia, and anemia (3). All patients had a complete clinical response. With a median follow-up of 17 mos (3-40 mos), no patients have had recurrence and all patients are still alive. Conclusions: This study demonstrates promising tolerability and efficacy for weekly C/P chemoradiation for patients with anal cancer unable to receive M/5FU. This regimen merits further investigation in prospective clinical trials.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...