User login
CVD Risk Rises With Higher NSAID Doses in Ankylosing Spondylitis
TOPLINE:
Higher doses of nonsteroidal anti-inflammatory drugs (NSAIDs) increase the risk for cardiovascular diseases (CVDs) such as ischemic heart disease, stroke, and congestive heart failure in patients with ankylosing spondylitis (AS) compared with lower doses.
METHODOLOGY:
- NSAIDs can suppress inflammation and relieve pain in patients with AS, but long-term treatment with NSAIDs poses concerns regarding gastrointestinal and renal toxicities and increased CVD risk.
- This nationwide cohort study used data from the Korean National Health Insurance database to investigate the risk for CVD associated with an increasing NSAID dosage in a real-world AS cohort.
- Investigators recruited 19,775 patients (mean age, 36.1 years; 75% men) with newly diagnosed AS and without any prior CVD between January 2010 and December 2018, among whom 99.7% received NSAID treatment and 30.2% received tumor necrosis factor inhibitor treatment.
- A time-varying approach was used to assess the NSAID exposure, wherein periods of NSAID use were defined as “NSAID-exposed” and periods longer than 1 month without NSAID use were defined as “NSAID-unexposed.”
- The primary outcome was the composite outcome of ischemic heart disease, stroke, or congestive heart failure.
TAKEAWAY:
- During the follow-up period of 98,290 person-years, 1663 cases of CVD were identified, which included 1157 cases of ischemic heart disease, 301 cases of stroke, and 613 cases of congestive heart failure.
- After adjusting for confounders, each defined daily dose increase in NSAIDs raised the risk for incident CVD by 10% (adjusted hazard ratio [aHR], 1.10; 95% CI, 1.08-1.13).
- Similarly, increasing the dose of NSAIDs was associated with an increased risk for ischemic heart disease (aHR, 1.08; 95% CI, 1.05-1.11), stroke (aHR, 1.09; 95% CI, 1.04-1.15), and congestive heart failure (aHR, 1.12; 95% CI, 1.08-1.16).
- The association between increasing NSAID dose and increased CVD risk was consistent across various subgroups, with NSAIDs posing a greater threat to cardiovascular health in women than in men.
IN PRACTICE:
The authors wrote, “Taken together, these results suggest that increasing the dose of NSAIDs is associated with a higher cardiovascular risk in AS, but that the increased risk might be lower than that in the general population.”
SOURCE:
First author Ji-Won Kim, MD, PhD, of the Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, the Republic of Korea, and colleagues had their work published online on April 9 in Annals of the Rheumatic Diseases.
LIMITATIONS:
The study was of retrospective nature. The levels of acute phase reactants and AS disease activity could not be determined owing to a lack of data in the National Health Insurance database. The accuracy of the diagnosis of cardiovascular outcomes on the basis of the International Classification of Disease codes was also questionable.
DISCLOSURES:
The study was supported by the National Research Foundation of Korea. The authors declared no conflicts of interest.
A version of this article appeared on Medscape.com.
TOPLINE:
Higher doses of nonsteroidal anti-inflammatory drugs (NSAIDs) increase the risk for cardiovascular diseases (CVDs) such as ischemic heart disease, stroke, and congestive heart failure in patients with ankylosing spondylitis (AS) compared with lower doses.
METHODOLOGY:
- NSAIDs can suppress inflammation and relieve pain in patients with AS, but long-term treatment with NSAIDs poses concerns regarding gastrointestinal and renal toxicities and increased CVD risk.
- This nationwide cohort study used data from the Korean National Health Insurance database to investigate the risk for CVD associated with an increasing NSAID dosage in a real-world AS cohort.
- Investigators recruited 19,775 patients (mean age, 36.1 years; 75% men) with newly diagnosed AS and without any prior CVD between January 2010 and December 2018, among whom 99.7% received NSAID treatment and 30.2% received tumor necrosis factor inhibitor treatment.
- A time-varying approach was used to assess the NSAID exposure, wherein periods of NSAID use were defined as “NSAID-exposed” and periods longer than 1 month without NSAID use were defined as “NSAID-unexposed.”
- The primary outcome was the composite outcome of ischemic heart disease, stroke, or congestive heart failure.
TAKEAWAY:
- During the follow-up period of 98,290 person-years, 1663 cases of CVD were identified, which included 1157 cases of ischemic heart disease, 301 cases of stroke, and 613 cases of congestive heart failure.
- After adjusting for confounders, each defined daily dose increase in NSAIDs raised the risk for incident CVD by 10% (adjusted hazard ratio [aHR], 1.10; 95% CI, 1.08-1.13).
- Similarly, increasing the dose of NSAIDs was associated with an increased risk for ischemic heart disease (aHR, 1.08; 95% CI, 1.05-1.11), stroke (aHR, 1.09; 95% CI, 1.04-1.15), and congestive heart failure (aHR, 1.12; 95% CI, 1.08-1.16).
- The association between increasing NSAID dose and increased CVD risk was consistent across various subgroups, with NSAIDs posing a greater threat to cardiovascular health in women than in men.
IN PRACTICE:
The authors wrote, “Taken together, these results suggest that increasing the dose of NSAIDs is associated with a higher cardiovascular risk in AS, but that the increased risk might be lower than that in the general population.”
SOURCE:
First author Ji-Won Kim, MD, PhD, of the Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, the Republic of Korea, and colleagues had their work published online on April 9 in Annals of the Rheumatic Diseases.
LIMITATIONS:
The study was of retrospective nature. The levels of acute phase reactants and AS disease activity could not be determined owing to a lack of data in the National Health Insurance database. The accuracy of the diagnosis of cardiovascular outcomes on the basis of the International Classification of Disease codes was also questionable.
DISCLOSURES:
The study was supported by the National Research Foundation of Korea. The authors declared no conflicts of interest.
A version of this article appeared on Medscape.com.
TOPLINE:
Higher doses of nonsteroidal anti-inflammatory drugs (NSAIDs) increase the risk for cardiovascular diseases (CVDs) such as ischemic heart disease, stroke, and congestive heart failure in patients with ankylosing spondylitis (AS) compared with lower doses.
METHODOLOGY:
- NSAIDs can suppress inflammation and relieve pain in patients with AS, but long-term treatment with NSAIDs poses concerns regarding gastrointestinal and renal toxicities and increased CVD risk.
- This nationwide cohort study used data from the Korean National Health Insurance database to investigate the risk for CVD associated with an increasing NSAID dosage in a real-world AS cohort.
- Investigators recruited 19,775 patients (mean age, 36.1 years; 75% men) with newly diagnosed AS and without any prior CVD between January 2010 and December 2018, among whom 99.7% received NSAID treatment and 30.2% received tumor necrosis factor inhibitor treatment.
- A time-varying approach was used to assess the NSAID exposure, wherein periods of NSAID use were defined as “NSAID-exposed” and periods longer than 1 month without NSAID use were defined as “NSAID-unexposed.”
- The primary outcome was the composite outcome of ischemic heart disease, stroke, or congestive heart failure.
TAKEAWAY:
- During the follow-up period of 98,290 person-years, 1663 cases of CVD were identified, which included 1157 cases of ischemic heart disease, 301 cases of stroke, and 613 cases of congestive heart failure.
- After adjusting for confounders, each defined daily dose increase in NSAIDs raised the risk for incident CVD by 10% (adjusted hazard ratio [aHR], 1.10; 95% CI, 1.08-1.13).
- Similarly, increasing the dose of NSAIDs was associated with an increased risk for ischemic heart disease (aHR, 1.08; 95% CI, 1.05-1.11), stroke (aHR, 1.09; 95% CI, 1.04-1.15), and congestive heart failure (aHR, 1.12; 95% CI, 1.08-1.16).
- The association between increasing NSAID dose and increased CVD risk was consistent across various subgroups, with NSAIDs posing a greater threat to cardiovascular health in women than in men.
IN PRACTICE:
The authors wrote, “Taken together, these results suggest that increasing the dose of NSAIDs is associated with a higher cardiovascular risk in AS, but that the increased risk might be lower than that in the general population.”
SOURCE:
First author Ji-Won Kim, MD, PhD, of the Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, the Republic of Korea, and colleagues had their work published online on April 9 in Annals of the Rheumatic Diseases.
LIMITATIONS:
The study was of retrospective nature. The levels of acute phase reactants and AS disease activity could not be determined owing to a lack of data in the National Health Insurance database. The accuracy of the diagnosis of cardiovascular outcomes on the basis of the International Classification of Disease codes was also questionable.
DISCLOSURES:
The study was supported by the National Research Foundation of Korea. The authors declared no conflicts of interest.
A version of this article appeared on Medscape.com.
Postpartum Depression
Improved Survival With Everolimus + Endocrine Therapy in HR+/HER2− Advanced BC
Key clinical point: Everolimus plus endocrine therapy (ET) led to promising survival outcomes in patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2−) advanced breast cancer (BC) who progressed on a CDK4/6 inhibitor.
Major finding: Everolimus + ET led to a median progression-free survival (mPFS) of 6 months (95% CI 5.3-7.8 months), with longer mPFS observed in patients previously treated with CDK4/6 inhibitors for >18 months (8.7 months; 95% CI 6.6-11.3 months), patients without visceral metastasis (8.0 months; 95% CI 5.8-10.5 months), and chemotherapy-naive patients (7.2 months; 95% CI 5.9-8.4 months).
Study details: This retrospective observational study included 161 patients with HR+/HER2− advanced BC who were previously treated with CDK4/6 inhibitors and received everolimus + ET.
Disclosures: This study did not receive any funding. Several authors declared receiving honoraria, research grants, or travel support from or having other ties with various sources.
Source: Sánchez-Bayona R, Lopez de Sa A, Jerez Gilarranz Y, et al. Everolimus plus endocrine therapy beyond CDK4/6 inhibitors progression for HR+ /HER2− advanced breast cancer: A real-world evidence cohort. Breast Cancer Res Treat. 2024 (May 4). doi: 10.1007/s10549-024-07324-8 Source
Key clinical point: Everolimus plus endocrine therapy (ET) led to promising survival outcomes in patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2−) advanced breast cancer (BC) who progressed on a CDK4/6 inhibitor.
Major finding: Everolimus + ET led to a median progression-free survival (mPFS) of 6 months (95% CI 5.3-7.8 months), with longer mPFS observed in patients previously treated with CDK4/6 inhibitors for >18 months (8.7 months; 95% CI 6.6-11.3 months), patients without visceral metastasis (8.0 months; 95% CI 5.8-10.5 months), and chemotherapy-naive patients (7.2 months; 95% CI 5.9-8.4 months).
Study details: This retrospective observational study included 161 patients with HR+/HER2− advanced BC who were previously treated with CDK4/6 inhibitors and received everolimus + ET.
Disclosures: This study did not receive any funding. Several authors declared receiving honoraria, research grants, or travel support from or having other ties with various sources.
Source: Sánchez-Bayona R, Lopez de Sa A, Jerez Gilarranz Y, et al. Everolimus plus endocrine therapy beyond CDK4/6 inhibitors progression for HR+ /HER2− advanced breast cancer: A real-world evidence cohort. Breast Cancer Res Treat. 2024 (May 4). doi: 10.1007/s10549-024-07324-8 Source
Key clinical point: Everolimus plus endocrine therapy (ET) led to promising survival outcomes in patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2−) advanced breast cancer (BC) who progressed on a CDK4/6 inhibitor.
Major finding: Everolimus + ET led to a median progression-free survival (mPFS) of 6 months (95% CI 5.3-7.8 months), with longer mPFS observed in patients previously treated with CDK4/6 inhibitors for >18 months (8.7 months; 95% CI 6.6-11.3 months), patients without visceral metastasis (8.0 months; 95% CI 5.8-10.5 months), and chemotherapy-naive patients (7.2 months; 95% CI 5.9-8.4 months).
Study details: This retrospective observational study included 161 patients with HR+/HER2− advanced BC who were previously treated with CDK4/6 inhibitors and received everolimus + ET.
Disclosures: This study did not receive any funding. Several authors declared receiving honoraria, research grants, or travel support from or having other ties with various sources.
Source: Sánchez-Bayona R, Lopez de Sa A, Jerez Gilarranz Y, et al. Everolimus plus endocrine therapy beyond CDK4/6 inhibitors progression for HR+ /HER2− advanced breast cancer: A real-world evidence cohort. Breast Cancer Res Treat. 2024 (May 4). doi: 10.1007/s10549-024-07324-8 Source
Anthracycline and Trastuzumab Tied to Long-Term CVD Risk in BC Survivors
Key clinical point: Chemotherapy with anthracycline or trastuzumab increased the risk for cardiovascular diseases (CVD) in breast cancer (BC) survivors, with the risk persisting beyond 10 years after BC diagnosis and being high in women age < 65 years.
Major finding: Anthracycline or trastuzumab vs no chemotherapy was associated with a 53% higher risk for incident CVD (adjusted hazard ratio [aHR] 1.53; 95% CI 1.31-1.79), particularly in women age < 65 years (aHR 1.70; 95% CI ≥1.19 to ≤2.45). The risk for CVD was seen beyond 5 years after BC diagnosis (aHR5-<10 years 1.85; 95% CI 1.44-2.39; aHR10+ years 1.83; 95% CI 1.34-2.49).
Study details: This retrospective cohort study included 10,211 female BC survivors who received anthracycline or trastuzumab (n = 2712), other chemotherapies (n = 1185), or no chemotherapy (n = 6314), with a median follow-up period of 5.79 years.
Disclosures: This study was supported in part by the Intramural Research Program of the US National Cancer Institute. The authors declared no conflicts of interest.
Source: Vo JB, Ramin C, Veiga LHS, et al. Long-term cardiovascular disease risk after anthracycline and trastuzumab treatments in U.S. breast cancer survivors. J Natl Cancer Inst. 2024 (May 8). doi: 10.1093/jnci/djae107 Source
Key clinical point: Chemotherapy with anthracycline or trastuzumab increased the risk for cardiovascular diseases (CVD) in breast cancer (BC) survivors, with the risk persisting beyond 10 years after BC diagnosis and being high in women age < 65 years.
Major finding: Anthracycline or trastuzumab vs no chemotherapy was associated with a 53% higher risk for incident CVD (adjusted hazard ratio [aHR] 1.53; 95% CI 1.31-1.79), particularly in women age < 65 years (aHR 1.70; 95% CI ≥1.19 to ≤2.45). The risk for CVD was seen beyond 5 years after BC diagnosis (aHR5-<10 years 1.85; 95% CI 1.44-2.39; aHR10+ years 1.83; 95% CI 1.34-2.49).
Study details: This retrospective cohort study included 10,211 female BC survivors who received anthracycline or trastuzumab (n = 2712), other chemotherapies (n = 1185), or no chemotherapy (n = 6314), with a median follow-up period of 5.79 years.
Disclosures: This study was supported in part by the Intramural Research Program of the US National Cancer Institute. The authors declared no conflicts of interest.
Source: Vo JB, Ramin C, Veiga LHS, et al. Long-term cardiovascular disease risk after anthracycline and trastuzumab treatments in U.S. breast cancer survivors. J Natl Cancer Inst. 2024 (May 8). doi: 10.1093/jnci/djae107 Source
Key clinical point: Chemotherapy with anthracycline or trastuzumab increased the risk for cardiovascular diseases (CVD) in breast cancer (BC) survivors, with the risk persisting beyond 10 years after BC diagnosis and being high in women age < 65 years.
Major finding: Anthracycline or trastuzumab vs no chemotherapy was associated with a 53% higher risk for incident CVD (adjusted hazard ratio [aHR] 1.53; 95% CI 1.31-1.79), particularly in women age < 65 years (aHR 1.70; 95% CI ≥1.19 to ≤2.45). The risk for CVD was seen beyond 5 years after BC diagnosis (aHR5-<10 years 1.85; 95% CI 1.44-2.39; aHR10+ years 1.83; 95% CI 1.34-2.49).
Study details: This retrospective cohort study included 10,211 female BC survivors who received anthracycline or trastuzumab (n = 2712), other chemotherapies (n = 1185), or no chemotherapy (n = 6314), with a median follow-up period of 5.79 years.
Disclosures: This study was supported in part by the Intramural Research Program of the US National Cancer Institute. The authors declared no conflicts of interest.
Source: Vo JB, Ramin C, Veiga LHS, et al. Long-term cardiovascular disease risk after anthracycline and trastuzumab treatments in U.S. breast cancer survivors. J Natl Cancer Inst. 2024 (May 8). doi: 10.1093/jnci/djae107 Source
Breast-Conserving Surgery Candidates Can Opt for Neoadjuvant Radiochemotherapy
Key clinical point: Neoadjuvant radiation therapy (NART) led to similar survival outcomes as postoperation radiation therapy (PORT) in patients with invasive ductal carcinoma (IDC) who underwent breast-conserving surgery (BCS) and were treated with neoadjuvant chemotherapy.
Major finding: NART vs PORT led to comparable breast cancer-specific survival (BCCS) and overall survival (OS) outcomes (both log-rank P > .05) in patients undergoing BCS or implant-based immediate breast reconstruction. However, NART vs PORT led to significantly lower BCCS (hazard ratio [HR] 1.407; log-rank P = .003) and OS (HR 1.383; log-rank P = .004) outcomes in those undergoing mastectomy.
Study details: This retrospective study included 14,515 women with IDC (age ≤ 80 years) from the Surveillance, Epidemiology, and End Results (SEER) database who were treated with neoadjuvant chemotherapy, of whom 386 and 14,129 patients underwent NART and PORT, respectively.
Disclosures: This study was supported by Shanghai Science and Technology Commission and Fudan University, China. The authors declared no conflicts of interest.
Source: Yuan J, Zhang M, Wang M, et al. Neoadjuvant radiochemotherapy is safe and feasible for breast conserving surgery or immediate reconstruction. Sci Rep. 2024;14:9208 (Apr 22). doi: 10.1038/s41598-024-59961-0 Source
Key clinical point: Neoadjuvant radiation therapy (NART) led to similar survival outcomes as postoperation radiation therapy (PORT) in patients with invasive ductal carcinoma (IDC) who underwent breast-conserving surgery (BCS) and were treated with neoadjuvant chemotherapy.
Major finding: NART vs PORT led to comparable breast cancer-specific survival (BCCS) and overall survival (OS) outcomes (both log-rank P > .05) in patients undergoing BCS or implant-based immediate breast reconstruction. However, NART vs PORT led to significantly lower BCCS (hazard ratio [HR] 1.407; log-rank P = .003) and OS (HR 1.383; log-rank P = .004) outcomes in those undergoing mastectomy.
Study details: This retrospective study included 14,515 women with IDC (age ≤ 80 years) from the Surveillance, Epidemiology, and End Results (SEER) database who were treated with neoadjuvant chemotherapy, of whom 386 and 14,129 patients underwent NART and PORT, respectively.
Disclosures: This study was supported by Shanghai Science and Technology Commission and Fudan University, China. The authors declared no conflicts of interest.
Source: Yuan J, Zhang M, Wang M, et al. Neoadjuvant radiochemotherapy is safe and feasible for breast conserving surgery or immediate reconstruction. Sci Rep. 2024;14:9208 (Apr 22). doi: 10.1038/s41598-024-59961-0 Source
Key clinical point: Neoadjuvant radiation therapy (NART) led to similar survival outcomes as postoperation radiation therapy (PORT) in patients with invasive ductal carcinoma (IDC) who underwent breast-conserving surgery (BCS) and were treated with neoadjuvant chemotherapy.
Major finding: NART vs PORT led to comparable breast cancer-specific survival (BCCS) and overall survival (OS) outcomes (both log-rank P > .05) in patients undergoing BCS or implant-based immediate breast reconstruction. However, NART vs PORT led to significantly lower BCCS (hazard ratio [HR] 1.407; log-rank P = .003) and OS (HR 1.383; log-rank P = .004) outcomes in those undergoing mastectomy.
Study details: This retrospective study included 14,515 women with IDC (age ≤ 80 years) from the Surveillance, Epidemiology, and End Results (SEER) database who were treated with neoadjuvant chemotherapy, of whom 386 and 14,129 patients underwent NART and PORT, respectively.
Disclosures: This study was supported by Shanghai Science and Technology Commission and Fudan University, China. The authors declared no conflicts of interest.
Source: Yuan J, Zhang M, Wang M, et al. Neoadjuvant radiochemotherapy is safe and feasible for breast conserving surgery or immediate reconstruction. Sci Rep. 2024;14:9208 (Apr 22). doi: 10.1038/s41598-024-59961-0 Source
Cancer Identified Via Serum Metabolites, Lipids in Rheumatic Disease or Paraneoplasia
TOPLINE:
A diagnostic model based on the concentrations of four metabolites and one lipid ratio can reliably predict cancer in patients with rheumatic and musculoskeletal diseases (RMDs) or paraneoplasia, providing high sensitivity and specificity.
METHODOLOGY:
- The metabolome profile can differentiate between nonspecific inflammatory symptoms such as those associated with paraneoplastic conditions or RMDs, which can help accelerate cancer diagnosis and treatment.
- To assess if changes in the serum metabolome profile could indicate cancer in patients with RMD, researchers performed nuclear magnetic resonance analysis of the sera of patients with rheumatoid arthritis (RA) with a history of invasive cancer (n = 56; age, 69.9 years; 76.8% women) or without such history (n = 52; age, 56.1 years; 57.7% women).
- Blinded validation was conducted in a cohort of patients with RA or spondyloarthritis with or without a history of invasive cancer.
- Additionally, the model performance was tested in a cohort of patients having RA or spondyloarthritis with active cancer or cancer treatment, pulmonary and lymphoid type cancers, paraneoplastic syndromes, and facultative solid noninvasive precancerous lesions and nonmelanoma skin cancer; in samples prior to the development of malignancy; and in a cohort of patients with systemic lupus erythematosus (SLE).
- The final model comprised five variables. The goodness of fit of the model was described using the area under the receiver operating characteristic curve (AUC).
TAKEAWAY:
- Based on the concentrations of acetate, creatine, glycine, and formate and the L1/L6 lipid ratio, the diagnostic model yielded an excellent AUC (0.987) and high sensitivity (0.932) and specificity (0.946) for cancer diagnosis in patients with RA.
- The diagnostic model yielded an AUC of 0.937 in the blinded validation cohort of patients with RA and an AUC of 0.927 in the merged RA and spondyloarthritis cohort.
- Although the diagnostic model accurately diagnosed cancer in all the patients with paraneoplasia, it could do so accurately in only 50% of patients with noninvasive or in situ precancerous lesions and nonmelanoma skin cancers.
- The performance of the model was poor in the SLE cohort (AUC, 0.656), and it could not identify patients at risk for later invasive cancer development.
IN PRACTICE:
“This limited-invasive assay has considerable potential of high clinical value to facilitate timely diagnosis of cancer in paraneoplastic rheumatic syndromes as well as become a valuable active surveillance tool in RA and SpA [spondyloarthritis] patients with a high risk of developing cancer,” the authors wrote.
SOURCE:
The study, led by Karolina Gente, MHBA, Heidelberg University Hospital, Heidelberg, Germany, was published online on April 1, 2024, in Annals of the Rheumatic Diseases.
LIMITATIONS:
The limited invasiveness during sampling might account for the model’s inability to identify three early-stage, low-grade tumors and its nonreliability in identifying noninvasive facultative precancerous lesions and nonmelanoma skin cancers. Given its poor performance in the SLE cohort, the model may not be suitable for universal application in more systemic rheumatic diseases.
DISCLOSURES:
This study was supported by an unrestricted investigator-initiated grant from the Foundation Commission of the Medical Faculty, University of Heidelberg, Germany. The authors declared no conflicts of interest.
A version of this article appeared on Medscape.com.
TOPLINE:
A diagnostic model based on the concentrations of four metabolites and one lipid ratio can reliably predict cancer in patients with rheumatic and musculoskeletal diseases (RMDs) or paraneoplasia, providing high sensitivity and specificity.
METHODOLOGY:
- The metabolome profile can differentiate between nonspecific inflammatory symptoms such as those associated with paraneoplastic conditions or RMDs, which can help accelerate cancer diagnosis and treatment.
- To assess if changes in the serum metabolome profile could indicate cancer in patients with RMD, researchers performed nuclear magnetic resonance analysis of the sera of patients with rheumatoid arthritis (RA) with a history of invasive cancer (n = 56; age, 69.9 years; 76.8% women) or without such history (n = 52; age, 56.1 years; 57.7% women).
- Blinded validation was conducted in a cohort of patients with RA or spondyloarthritis with or without a history of invasive cancer.
- Additionally, the model performance was tested in a cohort of patients having RA or spondyloarthritis with active cancer or cancer treatment, pulmonary and lymphoid type cancers, paraneoplastic syndromes, and facultative solid noninvasive precancerous lesions and nonmelanoma skin cancer; in samples prior to the development of malignancy; and in a cohort of patients with systemic lupus erythematosus (SLE).
- The final model comprised five variables. The goodness of fit of the model was described using the area under the receiver operating characteristic curve (AUC).
TAKEAWAY:
- Based on the concentrations of acetate, creatine, glycine, and formate and the L1/L6 lipid ratio, the diagnostic model yielded an excellent AUC (0.987) and high sensitivity (0.932) and specificity (0.946) for cancer diagnosis in patients with RA.
- The diagnostic model yielded an AUC of 0.937 in the blinded validation cohort of patients with RA and an AUC of 0.927 in the merged RA and spondyloarthritis cohort.
- Although the diagnostic model accurately diagnosed cancer in all the patients with paraneoplasia, it could do so accurately in only 50% of patients with noninvasive or in situ precancerous lesions and nonmelanoma skin cancers.
- The performance of the model was poor in the SLE cohort (AUC, 0.656), and it could not identify patients at risk for later invasive cancer development.
IN PRACTICE:
“This limited-invasive assay has considerable potential of high clinical value to facilitate timely diagnosis of cancer in paraneoplastic rheumatic syndromes as well as become a valuable active surveillance tool in RA and SpA [spondyloarthritis] patients with a high risk of developing cancer,” the authors wrote.
SOURCE:
The study, led by Karolina Gente, MHBA, Heidelberg University Hospital, Heidelberg, Germany, was published online on April 1, 2024, in Annals of the Rheumatic Diseases.
LIMITATIONS:
The limited invasiveness during sampling might account for the model’s inability to identify three early-stage, low-grade tumors and its nonreliability in identifying noninvasive facultative precancerous lesions and nonmelanoma skin cancers. Given its poor performance in the SLE cohort, the model may not be suitable for universal application in more systemic rheumatic diseases.
DISCLOSURES:
This study was supported by an unrestricted investigator-initiated grant from the Foundation Commission of the Medical Faculty, University of Heidelberg, Germany. The authors declared no conflicts of interest.
A version of this article appeared on Medscape.com.
TOPLINE:
A diagnostic model based on the concentrations of four metabolites and one lipid ratio can reliably predict cancer in patients with rheumatic and musculoskeletal diseases (RMDs) or paraneoplasia, providing high sensitivity and specificity.
METHODOLOGY:
- The metabolome profile can differentiate between nonspecific inflammatory symptoms such as those associated with paraneoplastic conditions or RMDs, which can help accelerate cancer diagnosis and treatment.
- To assess if changes in the serum metabolome profile could indicate cancer in patients with RMD, researchers performed nuclear magnetic resonance analysis of the sera of patients with rheumatoid arthritis (RA) with a history of invasive cancer (n = 56; age, 69.9 years; 76.8% women) or without such history (n = 52; age, 56.1 years; 57.7% women).
- Blinded validation was conducted in a cohort of patients with RA or spondyloarthritis with or without a history of invasive cancer.
- Additionally, the model performance was tested in a cohort of patients having RA or spondyloarthritis with active cancer or cancer treatment, pulmonary and lymphoid type cancers, paraneoplastic syndromes, and facultative solid noninvasive precancerous lesions and nonmelanoma skin cancer; in samples prior to the development of malignancy; and in a cohort of patients with systemic lupus erythematosus (SLE).
- The final model comprised five variables. The goodness of fit of the model was described using the area under the receiver operating characteristic curve (AUC).
TAKEAWAY:
- Based on the concentrations of acetate, creatine, glycine, and formate and the L1/L6 lipid ratio, the diagnostic model yielded an excellent AUC (0.987) and high sensitivity (0.932) and specificity (0.946) for cancer diagnosis in patients with RA.
- The diagnostic model yielded an AUC of 0.937 in the blinded validation cohort of patients with RA and an AUC of 0.927 in the merged RA and spondyloarthritis cohort.
- Although the diagnostic model accurately diagnosed cancer in all the patients with paraneoplasia, it could do so accurately in only 50% of patients with noninvasive or in situ precancerous lesions and nonmelanoma skin cancers.
- The performance of the model was poor in the SLE cohort (AUC, 0.656), and it could not identify patients at risk for later invasive cancer development.
IN PRACTICE:
“This limited-invasive assay has considerable potential of high clinical value to facilitate timely diagnosis of cancer in paraneoplastic rheumatic syndromes as well as become a valuable active surveillance tool in RA and SpA [spondyloarthritis] patients with a high risk of developing cancer,” the authors wrote.
SOURCE:
The study, led by Karolina Gente, MHBA, Heidelberg University Hospital, Heidelberg, Germany, was published online on April 1, 2024, in Annals of the Rheumatic Diseases.
LIMITATIONS:
The limited invasiveness during sampling might account for the model’s inability to identify three early-stage, low-grade tumors and its nonreliability in identifying noninvasive facultative precancerous lesions and nonmelanoma skin cancers. Given its poor performance in the SLE cohort, the model may not be suitable for universal application in more systemic rheumatic diseases.
DISCLOSURES:
This study was supported by an unrestricted investigator-initiated grant from the Foundation Commission of the Medical Faculty, University of Heidelberg, Germany. The authors declared no conflicts of interest.
A version of this article appeared on Medscape.com.
Molecular Classification of Endometrial Carcinomas
Historically, endometrial cancer has been classified as type I or type II. Type I endometrial cancers are typically estrogen driven, low grade, with endometrioid histology, and have a more favorable prognosis. In contrast, type II endometrial cancers are typically high grade, have more aggressive histologies (eg, serous or clear cell), and have a poorer prognosis.1
While this system provides a helpful schema for understanding endometrial cancers, it fails to represent the immense variation of biologic behavior and outcomes in endometrial cancers and oversimplifies what has come to be understood as a complex and molecularly diverse disease.
In 2013, The Cancer Genome Atlas (TCGA) performed genomic, transcriptomic, and proteomic characterization of 373 endometrial carcinomas. They identified four categories with distinct genetic profiles corresponding to clinical outcomes: 1) DNA polymerase epsilon (POLE) mutated; 2) mismatch repair deficient; 3) copy number high/p53 abnormal; and 4) copy number low/no specific molecular profile.2 By providing both predictive and prognostic information, these molecular features may be incorporated into treatment planning decisions in the future.
The POLE-mutated subtype are endometrial cancers with recurrent mutations in the POLE gene, which is involved in DNA replication and repair. POLE mutations occur in about 5%-10% of endometrial cancers. Despite some more aggressive histopathologic findings (eg, higher grade, deeper myometrial invasion, positive lymphovascular space invasion), recurrences rarely occur, and patients with POLE mutations have the best prognosis of the four molecular subtypes, with a 5-year recurrence-free survival of 92%-100%.3
The mismatch repair–deficient (MMRd) subtype are endometrial cancers with abnormalities in any of the mismatch repair proteins (MLH1, PMS2, MSH2, MSH6). These alterations may result from hereditary or somatic mutations in any of the MMR genes or epigenetic changes in the MLH1 promoter. Germ-line mutations are associated with Lynch syndrome; thus, patients found to have a germ-line mutation in any of the MMR genes necessitate a genetics referral. The MMRd subtype accounts for about 20%-30% of endometrial cancers, and patients with MMRd tumors have an intermediate prognosis, with a 5-year recurrence-free survival of about 70%.3. These tumors are more responsive to the use of immunotherapy checkpoint inhibitors. Two recent landmark trials showed improved outcomes in patients with advanced MMRd endometrial cancer treated with immune checkpoint inhibitors in addition to standard chemotherapy.4,5
The worst prognosis belongs to the copy number high subgroup, which accounts for approximately 10% of endometrial cancers. Five-year recurrence-free survival is ~50%.3 These tumors often contain TP53 mutations and are composed of aggressive histologies, such as serous, clear cell, high-grade endometrioid, and carcinosarcomas. Recent data suggests that human epidermal growth factor receptor 2 (HER2) amplification may also be prevalent in this subgroup.6
Endometrial cancers that lack any of the above alterations fall into the no specific molecular profile (NSMP) or copy number low subgroup. Mutations in other genes, such as PTEN, PIK3CA, CTNNB1, KRAS, and ARID1A, are often present in these tumors. As the most common subtype, this heterogeneous group accounts for about 50% of all endometrial cancers. These tumors frequently comprise endometrioid histology with estrogen and progesterone receptor positivity, high rates of response to hormonal therapy, and an overall intermediate to favorable prognosis, with a 5-year recurrence-free survival of ~75%.3
The use of whole-genome sequencing in TCGA limits the clinical applicability of testing because of the cost and complex methodologies involved. Multiple algorithms have been developed in the interim that approximate TCGA subtypes using relatively less expensive and more widely available testing methods, such as immunohistochemistry and next-generation sequencing. In the ProMisE algorithm, immunohistochemistry for p53 and MMR proteins is used as a surrogate for copy number high and MMRd tumors, respectively, and targeted sequencing is used to identify POLE mutations.7
Full molecular classification of endometrial tumors provides important prognostic information and allows for incorporation into treatment planning. To this end, the new 2023 International Federation of Gynecology and Obstetrics (FIGO) endometrial cancer staging incorporates an option for the addition of molecular subtype, with the stance that it allows for better prognostic prediction.8 While complete molecular classification is not required, it is encouraged. Furthermore, several clinical trials are currently investigating different treatment regimens based on these distinct molecular profiles.
Dr. Haag is a gynecologic oncology fellow in the Department of Obstetrics and Gynecology, University of North Carolina Hospitals, Chapel Hill. Dr. Tucker is assistant professor of gynecologic oncology at the University of North Carolina at Chapel Hill. They have no conflicts of interest.
References
1. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecologic Oncology. 1983;15(1):10-17.
2. Kandoth C et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67-73.
3. León-Castillo A et al. Molecular classification of the PORTEC-3 trial for high-risk endometrial cancer: Impact on prognosis and benefit from adjuvant therapy. J Clin Oncology. 2020;38(29):3388-3397.
4. Mirza MR et al. Dostarlimab for primary advanced or recurrent endometrial cancer. N Engl J Med. 2023;388(23):2145-2158.
5. Eskander RN et al. Pembrolizumab plus chemotherapy in advanced endometrial cancer. N Engl J Med. 2023;388(23):2159-2170.
6. Talia KL et al. The role of HER2 as a therapeutic biomarker in gynaecological malignancy: Potential for use beyond uterine serous carcinoma. Pathology. 2023;55(1):8-18.
7. Kommoss S et al. Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. Annals Oncology. 2018;29(5):1180-1188.
8. Berek JS et al. FIGO staging of endometrial cancer: 2023. Int J Gynaecol Obstet. 2023;162(2):383-394.
Historically, endometrial cancer has been classified as type I or type II. Type I endometrial cancers are typically estrogen driven, low grade, with endometrioid histology, and have a more favorable prognosis. In contrast, type II endometrial cancers are typically high grade, have more aggressive histologies (eg, serous or clear cell), and have a poorer prognosis.1
While this system provides a helpful schema for understanding endometrial cancers, it fails to represent the immense variation of biologic behavior and outcomes in endometrial cancers and oversimplifies what has come to be understood as a complex and molecularly diverse disease.
In 2013, The Cancer Genome Atlas (TCGA) performed genomic, transcriptomic, and proteomic characterization of 373 endometrial carcinomas. They identified four categories with distinct genetic profiles corresponding to clinical outcomes: 1) DNA polymerase epsilon (POLE) mutated; 2) mismatch repair deficient; 3) copy number high/p53 abnormal; and 4) copy number low/no specific molecular profile.2 By providing both predictive and prognostic information, these molecular features may be incorporated into treatment planning decisions in the future.
The POLE-mutated subtype are endometrial cancers with recurrent mutations in the POLE gene, which is involved in DNA replication and repair. POLE mutations occur in about 5%-10% of endometrial cancers. Despite some more aggressive histopathologic findings (eg, higher grade, deeper myometrial invasion, positive lymphovascular space invasion), recurrences rarely occur, and patients with POLE mutations have the best prognosis of the four molecular subtypes, with a 5-year recurrence-free survival of 92%-100%.3
The mismatch repair–deficient (MMRd) subtype are endometrial cancers with abnormalities in any of the mismatch repair proteins (MLH1, PMS2, MSH2, MSH6). These alterations may result from hereditary or somatic mutations in any of the MMR genes or epigenetic changes in the MLH1 promoter. Germ-line mutations are associated with Lynch syndrome; thus, patients found to have a germ-line mutation in any of the MMR genes necessitate a genetics referral. The MMRd subtype accounts for about 20%-30% of endometrial cancers, and patients with MMRd tumors have an intermediate prognosis, with a 5-year recurrence-free survival of about 70%.3. These tumors are more responsive to the use of immunotherapy checkpoint inhibitors. Two recent landmark trials showed improved outcomes in patients with advanced MMRd endometrial cancer treated with immune checkpoint inhibitors in addition to standard chemotherapy.4,5
The worst prognosis belongs to the copy number high subgroup, which accounts for approximately 10% of endometrial cancers. Five-year recurrence-free survival is ~50%.3 These tumors often contain TP53 mutations and are composed of aggressive histologies, such as serous, clear cell, high-grade endometrioid, and carcinosarcomas. Recent data suggests that human epidermal growth factor receptor 2 (HER2) amplification may also be prevalent in this subgroup.6
Endometrial cancers that lack any of the above alterations fall into the no specific molecular profile (NSMP) or copy number low subgroup. Mutations in other genes, such as PTEN, PIK3CA, CTNNB1, KRAS, and ARID1A, are often present in these tumors. As the most common subtype, this heterogeneous group accounts for about 50% of all endometrial cancers. These tumors frequently comprise endometrioid histology with estrogen and progesterone receptor positivity, high rates of response to hormonal therapy, and an overall intermediate to favorable prognosis, with a 5-year recurrence-free survival of ~75%.3
The use of whole-genome sequencing in TCGA limits the clinical applicability of testing because of the cost and complex methodologies involved. Multiple algorithms have been developed in the interim that approximate TCGA subtypes using relatively less expensive and more widely available testing methods, such as immunohistochemistry and next-generation sequencing. In the ProMisE algorithm, immunohistochemistry for p53 and MMR proteins is used as a surrogate for copy number high and MMRd tumors, respectively, and targeted sequencing is used to identify POLE mutations.7
Full molecular classification of endometrial tumors provides important prognostic information and allows for incorporation into treatment planning. To this end, the new 2023 International Federation of Gynecology and Obstetrics (FIGO) endometrial cancer staging incorporates an option for the addition of molecular subtype, with the stance that it allows for better prognostic prediction.8 While complete molecular classification is not required, it is encouraged. Furthermore, several clinical trials are currently investigating different treatment regimens based on these distinct molecular profiles.
Dr. Haag is a gynecologic oncology fellow in the Department of Obstetrics and Gynecology, University of North Carolina Hospitals, Chapel Hill. Dr. Tucker is assistant professor of gynecologic oncology at the University of North Carolina at Chapel Hill. They have no conflicts of interest.
References
1. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecologic Oncology. 1983;15(1):10-17.
2. Kandoth C et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67-73.
3. León-Castillo A et al. Molecular classification of the PORTEC-3 trial for high-risk endometrial cancer: Impact on prognosis and benefit from adjuvant therapy. J Clin Oncology. 2020;38(29):3388-3397.
4. Mirza MR et al. Dostarlimab for primary advanced or recurrent endometrial cancer. N Engl J Med. 2023;388(23):2145-2158.
5. Eskander RN et al. Pembrolizumab plus chemotherapy in advanced endometrial cancer. N Engl J Med. 2023;388(23):2159-2170.
6. Talia KL et al. The role of HER2 as a therapeutic biomarker in gynaecological malignancy: Potential for use beyond uterine serous carcinoma. Pathology. 2023;55(1):8-18.
7. Kommoss S et al. Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. Annals Oncology. 2018;29(5):1180-1188.
8. Berek JS et al. FIGO staging of endometrial cancer: 2023. Int J Gynaecol Obstet. 2023;162(2):383-394.
Historically, endometrial cancer has been classified as type I or type II. Type I endometrial cancers are typically estrogen driven, low grade, with endometrioid histology, and have a more favorable prognosis. In contrast, type II endometrial cancers are typically high grade, have more aggressive histologies (eg, serous or clear cell), and have a poorer prognosis.1
While this system provides a helpful schema for understanding endometrial cancers, it fails to represent the immense variation of biologic behavior and outcomes in endometrial cancers and oversimplifies what has come to be understood as a complex and molecularly diverse disease.
In 2013, The Cancer Genome Atlas (TCGA) performed genomic, transcriptomic, and proteomic characterization of 373 endometrial carcinomas. They identified four categories with distinct genetic profiles corresponding to clinical outcomes: 1) DNA polymerase epsilon (POLE) mutated; 2) mismatch repair deficient; 3) copy number high/p53 abnormal; and 4) copy number low/no specific molecular profile.2 By providing both predictive and prognostic information, these molecular features may be incorporated into treatment planning decisions in the future.
The POLE-mutated subtype are endometrial cancers with recurrent mutations in the POLE gene, which is involved in DNA replication and repair. POLE mutations occur in about 5%-10% of endometrial cancers. Despite some more aggressive histopathologic findings (eg, higher grade, deeper myometrial invasion, positive lymphovascular space invasion), recurrences rarely occur, and patients with POLE mutations have the best prognosis of the four molecular subtypes, with a 5-year recurrence-free survival of 92%-100%.3
The mismatch repair–deficient (MMRd) subtype are endometrial cancers with abnormalities in any of the mismatch repair proteins (MLH1, PMS2, MSH2, MSH6). These alterations may result from hereditary or somatic mutations in any of the MMR genes or epigenetic changes in the MLH1 promoter. Germ-line mutations are associated with Lynch syndrome; thus, patients found to have a germ-line mutation in any of the MMR genes necessitate a genetics referral. The MMRd subtype accounts for about 20%-30% of endometrial cancers, and patients with MMRd tumors have an intermediate prognosis, with a 5-year recurrence-free survival of about 70%.3. These tumors are more responsive to the use of immunotherapy checkpoint inhibitors. Two recent landmark trials showed improved outcomes in patients with advanced MMRd endometrial cancer treated with immune checkpoint inhibitors in addition to standard chemotherapy.4,5
The worst prognosis belongs to the copy number high subgroup, which accounts for approximately 10% of endometrial cancers. Five-year recurrence-free survival is ~50%.3 These tumors often contain TP53 mutations and are composed of aggressive histologies, such as serous, clear cell, high-grade endometrioid, and carcinosarcomas. Recent data suggests that human epidermal growth factor receptor 2 (HER2) amplification may also be prevalent in this subgroup.6
Endometrial cancers that lack any of the above alterations fall into the no specific molecular profile (NSMP) or copy number low subgroup. Mutations in other genes, such as PTEN, PIK3CA, CTNNB1, KRAS, and ARID1A, are often present in these tumors. As the most common subtype, this heterogeneous group accounts for about 50% of all endometrial cancers. These tumors frequently comprise endometrioid histology with estrogen and progesterone receptor positivity, high rates of response to hormonal therapy, and an overall intermediate to favorable prognosis, with a 5-year recurrence-free survival of ~75%.3
The use of whole-genome sequencing in TCGA limits the clinical applicability of testing because of the cost and complex methodologies involved. Multiple algorithms have been developed in the interim that approximate TCGA subtypes using relatively less expensive and more widely available testing methods, such as immunohistochemistry and next-generation sequencing. In the ProMisE algorithm, immunohistochemistry for p53 and MMR proteins is used as a surrogate for copy number high and MMRd tumors, respectively, and targeted sequencing is used to identify POLE mutations.7
Full molecular classification of endometrial tumors provides important prognostic information and allows for incorporation into treatment planning. To this end, the new 2023 International Federation of Gynecology and Obstetrics (FIGO) endometrial cancer staging incorporates an option for the addition of molecular subtype, with the stance that it allows for better prognostic prediction.8 While complete molecular classification is not required, it is encouraged. Furthermore, several clinical trials are currently investigating different treatment regimens based on these distinct molecular profiles.
Dr. Haag is a gynecologic oncology fellow in the Department of Obstetrics and Gynecology, University of North Carolina Hospitals, Chapel Hill. Dr. Tucker is assistant professor of gynecologic oncology at the University of North Carolina at Chapel Hill. They have no conflicts of interest.
References
1. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecologic Oncology. 1983;15(1):10-17.
2. Kandoth C et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67-73.
3. León-Castillo A et al. Molecular classification of the PORTEC-3 trial for high-risk endometrial cancer: Impact on prognosis and benefit from adjuvant therapy. J Clin Oncology. 2020;38(29):3388-3397.
4. Mirza MR et al. Dostarlimab for primary advanced or recurrent endometrial cancer. N Engl J Med. 2023;388(23):2145-2158.
5. Eskander RN et al. Pembrolizumab plus chemotherapy in advanced endometrial cancer. N Engl J Med. 2023;388(23):2159-2170.
6. Talia KL et al. The role of HER2 as a therapeutic biomarker in gynaecological malignancy: Potential for use beyond uterine serous carcinoma. Pathology. 2023;55(1):8-18.
7. Kommoss S et al. Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. Annals Oncology. 2018;29(5):1180-1188.
8. Berek JS et al. FIGO staging of endometrial cancer: 2023. Int J Gynaecol Obstet. 2023;162(2):383-394.
Statin Use Improves Cancer-Specific Survival in Older Women With Breast Cancer
Key clinical point: Use of a statin after breast cancer (BC) diagnosis improved survival in older women (age ≥ 66 years) with localized and regional stage disease, particularly in those with the hormone receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2−) subtype.
Major finding: Use vs no use of a statin postdiagnosis was associated with a 15% reduced risk for BC-specific mortality (hazard ratio 0.85; 95% CI 0.75-0.96), with the effect being more pronounced women with HR+/HER2− BC (hazard ratio 0.71; 95% CI 0.57-0.88). There was no significant association between postdiagnosis statin use and the risk for BC recurrence (hazard ratio 1.05; 95% CI 0.91-1.21).
Study details: This retrospective cohort study included women with localized and regional stage BC from the Surveillance, Epidemiology, and End Results (SEER)–Medicare database who were assessed for mortality (n = 38,858) and recurrence (n = 28,522), of whom 8836 and 6475 used a statin postdiagnosis, respectively.
Disclosures: This study was supported by the US National Cancer Institute, National Institutes of Health. The authors declared no conflicts of interest.
Source: Guo H, Malone KE, Heckbert SR, Li CI. Statin use and risks of breast cancer recurrence and mortality. Cancer. 2024 (May 6). doi: 10.1002/cncr.35362 Source
Key clinical point: Use of a statin after breast cancer (BC) diagnosis improved survival in older women (age ≥ 66 years) with localized and regional stage disease, particularly in those with the hormone receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2−) subtype.
Major finding: Use vs no use of a statin postdiagnosis was associated with a 15% reduced risk for BC-specific mortality (hazard ratio 0.85; 95% CI 0.75-0.96), with the effect being more pronounced women with HR+/HER2− BC (hazard ratio 0.71; 95% CI 0.57-0.88). There was no significant association between postdiagnosis statin use and the risk for BC recurrence (hazard ratio 1.05; 95% CI 0.91-1.21).
Study details: This retrospective cohort study included women with localized and regional stage BC from the Surveillance, Epidemiology, and End Results (SEER)–Medicare database who were assessed for mortality (n = 38,858) and recurrence (n = 28,522), of whom 8836 and 6475 used a statin postdiagnosis, respectively.
Disclosures: This study was supported by the US National Cancer Institute, National Institutes of Health. The authors declared no conflicts of interest.
Source: Guo H, Malone KE, Heckbert SR, Li CI. Statin use and risks of breast cancer recurrence and mortality. Cancer. 2024 (May 6). doi: 10.1002/cncr.35362 Source
Key clinical point: Use of a statin after breast cancer (BC) diagnosis improved survival in older women (age ≥ 66 years) with localized and regional stage disease, particularly in those with the hormone receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2−) subtype.
Major finding: Use vs no use of a statin postdiagnosis was associated with a 15% reduced risk for BC-specific mortality (hazard ratio 0.85; 95% CI 0.75-0.96), with the effect being more pronounced women with HR+/HER2− BC (hazard ratio 0.71; 95% CI 0.57-0.88). There was no significant association between postdiagnosis statin use and the risk for BC recurrence (hazard ratio 1.05; 95% CI 0.91-1.21).
Study details: This retrospective cohort study included women with localized and regional stage BC from the Surveillance, Epidemiology, and End Results (SEER)–Medicare database who were assessed for mortality (n = 38,858) and recurrence (n = 28,522), of whom 8836 and 6475 used a statin postdiagnosis, respectively.
Disclosures: This study was supported by the US National Cancer Institute, National Institutes of Health. The authors declared no conflicts of interest.
Source: Guo H, Malone KE, Heckbert SR, Li CI. Statin use and risks of breast cancer recurrence and mortality. Cancer. 2024 (May 6). doi: 10.1002/cncr.35362 Source
SLNB Not Required Before Neoadjuvant Chemotherapy in Node-Negative Breast Cancer
Key clinical point: Sentinel lymph node biopsy (SLNB) performed before vs after neoadjuvant chemotherapy (NACT) showed a higher axillary lymph node dissection rate and no overall survival (OS) benefits in patients with clinically lymph node-negative (cN0) breast cancer (BC).
Major finding: The axillary lymph node dissection rate was significantly higher for SLNB performed before vs after NACT (29.9% vs 7.4%; P < .001; odds ratio 5.35; P = .002). Moreover, the 4-year overall survival rate was significantly compromised when SLNB was performed before vs after NACT (88.4% vs 95.7%; hazard ratio 0.21; P = .009).
Study details: This retrospective observational study included 310 patients with cN0 BC, of whom 107 and 203 patients underwent SLNB before and after NACT, respectively.
Disclosures: This study did not receive any specific funding except Open Access funding from Springer Nature. The authors declared no financial conflicts of interest. Two authors declared non-financial ties with various sources.
Source: Fernandez-Gonzalez S, Falo C, Pla MJ, et al. Sentinel lymph node biopsy before and after neoadjuvant chemotherapy in cN0 breast cancer patients: Impact on axillary morbidity and survival—A propensity score cohort study. Breast Cancer Res Treat. 2024 (Apr 18). doi: 10.1007/s10549-024-07274-1 Source
Key clinical point: Sentinel lymph node biopsy (SLNB) performed before vs after neoadjuvant chemotherapy (NACT) showed a higher axillary lymph node dissection rate and no overall survival (OS) benefits in patients with clinically lymph node-negative (cN0) breast cancer (BC).
Major finding: The axillary lymph node dissection rate was significantly higher for SLNB performed before vs after NACT (29.9% vs 7.4%; P < .001; odds ratio 5.35; P = .002). Moreover, the 4-year overall survival rate was significantly compromised when SLNB was performed before vs after NACT (88.4% vs 95.7%; hazard ratio 0.21; P = .009).
Study details: This retrospective observational study included 310 patients with cN0 BC, of whom 107 and 203 patients underwent SLNB before and after NACT, respectively.
Disclosures: This study did not receive any specific funding except Open Access funding from Springer Nature. The authors declared no financial conflicts of interest. Two authors declared non-financial ties with various sources.
Source: Fernandez-Gonzalez S, Falo C, Pla MJ, et al. Sentinel lymph node biopsy before and after neoadjuvant chemotherapy in cN0 breast cancer patients: Impact on axillary morbidity and survival—A propensity score cohort study. Breast Cancer Res Treat. 2024 (Apr 18). doi: 10.1007/s10549-024-07274-1 Source
Key clinical point: Sentinel lymph node biopsy (SLNB) performed before vs after neoadjuvant chemotherapy (NACT) showed a higher axillary lymph node dissection rate and no overall survival (OS) benefits in patients with clinically lymph node-negative (cN0) breast cancer (BC).
Major finding: The axillary lymph node dissection rate was significantly higher for SLNB performed before vs after NACT (29.9% vs 7.4%; P < .001; odds ratio 5.35; P = .002). Moreover, the 4-year overall survival rate was significantly compromised when SLNB was performed before vs after NACT (88.4% vs 95.7%; hazard ratio 0.21; P = .009).
Study details: This retrospective observational study included 310 patients with cN0 BC, of whom 107 and 203 patients underwent SLNB before and after NACT, respectively.
Disclosures: This study did not receive any specific funding except Open Access funding from Springer Nature. The authors declared no financial conflicts of interest. Two authors declared non-financial ties with various sources.
Source: Fernandez-Gonzalez S, Falo C, Pla MJ, et al. Sentinel lymph node biopsy before and after neoadjuvant chemotherapy in cN0 breast cancer patients: Impact on axillary morbidity and survival—A propensity score cohort study. Breast Cancer Res Treat. 2024 (Apr 18). doi: 10.1007/s10549-024-07274-1 Source
Trastuzumab Deruxtecan Bests Trastuzumab Emtansine in HER2+ BC With or Without Brain Metastases
Key clinical point: Trastuzumab deruxtecan demonstrated superior efficacy over trastuzumab emtansine as second-line treatment in patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC) with or without brain metastases.
Major finding: Trastuzumab deruxtecan led to significantly longer median progression-free survival (15.0 vs 3.0 months; hazard ratio 0.25; 95% CI 0.13-0.45) and higher systemic (67.4% vs 20.5%) and intracranial (65.7% vs 34.3%) objective response rates than trastuzumab emtansine in patients with brain metastases. Outcomes were similar in patients without brain metastases.
Study details: This exploratory analysis of the phase 3 DESTINY-Breast03 trial included 524 patients with HER2+ metastatic BC with or without brain metastases who were randomly assigned to receive trastuzumab deruxtecan or trastuzumab emtansine after their disease progressed with trastuzumab and taxane treatment.
Disclosures: This study was supported by Daiichi Sankyo and AstraZeneca. Six authors declared being current or former employees or holding stock or stock options of Daiichi Sankyo or AstraZeneca. Several authors declared having ties to various sources, including Daiichi Sankyo and AstraZeneca.
Source: Hurvitz SA, Kim SB, Chung WP, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in HER2-positive metastatic breast cancer patients with brain metastases from the randomized DESTINY-Breast03 trial. ESMO Open. 2024;109294 (Apr 24). doi: 10.1016/j.esmoop.2024.102924 Source
Key clinical point: Trastuzumab deruxtecan demonstrated superior efficacy over trastuzumab emtansine as second-line treatment in patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC) with or without brain metastases.
Major finding: Trastuzumab deruxtecan led to significantly longer median progression-free survival (15.0 vs 3.0 months; hazard ratio 0.25; 95% CI 0.13-0.45) and higher systemic (67.4% vs 20.5%) and intracranial (65.7% vs 34.3%) objective response rates than trastuzumab emtansine in patients with brain metastases. Outcomes were similar in patients without brain metastases.
Study details: This exploratory analysis of the phase 3 DESTINY-Breast03 trial included 524 patients with HER2+ metastatic BC with or without brain metastases who were randomly assigned to receive trastuzumab deruxtecan or trastuzumab emtansine after their disease progressed with trastuzumab and taxane treatment.
Disclosures: This study was supported by Daiichi Sankyo and AstraZeneca. Six authors declared being current or former employees or holding stock or stock options of Daiichi Sankyo or AstraZeneca. Several authors declared having ties to various sources, including Daiichi Sankyo and AstraZeneca.
Source: Hurvitz SA, Kim SB, Chung WP, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in HER2-positive metastatic breast cancer patients with brain metastases from the randomized DESTINY-Breast03 trial. ESMO Open. 2024;109294 (Apr 24). doi: 10.1016/j.esmoop.2024.102924 Source
Key clinical point: Trastuzumab deruxtecan demonstrated superior efficacy over trastuzumab emtansine as second-line treatment in patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC) with or without brain metastases.
Major finding: Trastuzumab deruxtecan led to significantly longer median progression-free survival (15.0 vs 3.0 months; hazard ratio 0.25; 95% CI 0.13-0.45) and higher systemic (67.4% vs 20.5%) and intracranial (65.7% vs 34.3%) objective response rates than trastuzumab emtansine in patients with brain metastases. Outcomes were similar in patients without brain metastases.
Study details: This exploratory analysis of the phase 3 DESTINY-Breast03 trial included 524 patients with HER2+ metastatic BC with or without brain metastases who were randomly assigned to receive trastuzumab deruxtecan or trastuzumab emtansine after their disease progressed with trastuzumab and taxane treatment.
Disclosures: This study was supported by Daiichi Sankyo and AstraZeneca. Six authors declared being current or former employees or holding stock or stock options of Daiichi Sankyo or AstraZeneca. Several authors declared having ties to various sources, including Daiichi Sankyo and AstraZeneca.
Source: Hurvitz SA, Kim SB, Chung WP, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in HER2-positive metastatic breast cancer patients with brain metastases from the randomized DESTINY-Breast03 trial. ESMO Open. 2024;109294 (Apr 24). doi: 10.1016/j.esmoop.2024.102924 Source