Open access
Open access
Powered by Google Translator Translator

Oncology (all articles)

Phase III RCT: Adding Nab-Paclitaxel to Gemcitabine-Cisplatin Fails to Improve Survival in Advanced Biliary Tract Cancers

8 Jan, 2025 | 13:00h | UTC

Background: Biliary tract cancers (BTCs)—including intrahepatic and extrahepatic cholangiocarcinomas and gallbladder carcinoma—are often diagnosed at advanced stages, leaving few curative options. Gemcitabine-cisplatin (GC) has been the longstanding frontline therapy, yielding modest survival benefits. Preclinical research suggested that nab-paclitaxel might enhance drug delivery to desmoplastic tumors, and an encouraging phase II trial (GAP: gemcitabine, nab-paclitaxel, and cisplatin) spurred further investigation.

Objective: SWOG S1815 was designed to determine whether adding nab-paclitaxel (GAP regimen) to standard GC therapy would improve overall survival (OS) for patients newly diagnosed with locally advanced or metastatic BTCs.

Methods: This phase III, randomized, open-label trial enrolled patients with histologically or cytologically confirmed advanced BTC, excluding those with ampullary cancer (no prior systemic therapy for advanced disease). Participants were randomized 2:1 to receive either GAP (gemcitabine 800 mg/m^2, cisplatin 25 mg/m^2, nab-paclitaxel 100 mg/m^2 on days 1 and 8 of a 21-day cycle) or standard GC (gemcitabine 1,000 mg/m^2 plus cisplatin 25 mg/m^2 on days 1 and 8). The primary outcome was OS; secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and toxicity. Disease site (intrahepatic cholangiocarcinoma, extrahepatic cholangiocarcinoma, gallbladder carcinoma) and stage (locally advanced or metastatic) were key stratification factors.

Results: Among 441 eligible patients, 67% had intrahepatic cholangiocarcinoma, 16% had gallbladder carcinoma, and 17% had extrahepatic cholangiocarcinoma, with 73% presenting metastatic disease. Median OS was 14.0 months (95% confidence interval [CI], 12.4-16.1) in the GAP arm versus 13.6 months (95% CI, 9.7-16.6) in the GC arm (hazard ratio [HR], 0.91; P = .41). Similarly, median PFS did not significantly differ (7.5 months v 6.3 months; HR, 0.89; P = .32). Subset analyses hinted that patients with gallbladder carcinoma and those with locally advanced disease might benefit more from the GAP triplet, but these findings were not statistically conclusive. Although ORR was higher with GAP (31% v 21%; P = .03), this did not translate into improved OS. Treatment-related toxicities were more frequent with the triplet regimen. Grade 3 or higher hematologic adverse events occurred in 60% of GAP patients versus 45% of those on GC (P = .003). Additionally, several grade 3-4 nonhematologic toxicities (such as ALT increase, anorexia, constipation, diarrhea, edema, fatigue, hypomagnesemia, nausea, sepsis, sensory peripheral neuropathy, and vomiting) were significantly more common with GAP. Seven treatment-related deaths occurred in the GAP arm, versus one in the GC arm, underscoring the regimen’s higher toxicity burden.

Conclusions: In this unselected BTC population, adding nab-paclitaxel to standard gemcitabine-cisplatin did not confer an OS advantage. Although higher ORR was observed, the toxicity profile was notably increased. Based on these findings, adding nab-paclitaxel to GC does not improve OS and is associated with increased toxicity. Therefore, GC remains the standard first-line treatment for advanced BTC. However, similar to what was observed in the PRODIGE 38 AMEBICA trial, which compared mFOLFIRINOX to GC, there was no survival advantage with the triplet regimen.

Implications for Practice: From a real-world perspective, the triplet regimen elevates both toxicity and likely treatment costs, without demonstrable survival benefit for the broader BTC population. Clinicians may consider GAP in highly selected subsets (for instance, localized unresectable gallbladder disease) or in research settings. Until further evidence clarifies which patients might benefit, GC remains the standard first-line treatment for advanced BTC.

Study Strengths and Limitations: Strengths include the large, multi-institutional, US-based randomized design and robust accrual (enrollment of a sufficient number of participants).* Limitations involve the heterogeneous nature (diversity) of BTCs and the absence of centralized radiologic review (a process where imaging studies are reviewed by a central group of experts rather than solely by local investigators). Furthermore, the trial did not incorporate cost-effectiveness analyses or routine genomic stratification *(classifying patients based on the genetic characteristics of their tumors), which could have refined patient selection.

Future Research: Ongoing investigations seek to integrate precision oncology—encompassing genetic profiling and circulating tumor DNA—to identify subgroups that may benefit from targeted or intensified cytotoxic strategies. Perioperative or adjuvant approaches with GAP or similar triplets might prove more effective in earlier-stage disease. Studies combining novel immunotherapies or targeted agents with chemotherapy could address the evolving BTC landscape.

Reference: Shroff RT, King G, Colby S, et al. SWOG S1815: A Phase III Randomized Trial of Gemcitabine, Cisplatin, and Nab-Paclitaxel Versus Gemcitabine and Cisplatin in Newly Diagnosed, Advanced Biliary Tract Cancers. Journal of Clinical Oncology.
DOI: https://doi.org/10.1200/JCO-24-01383


RCT: Camrelizumab Increases Pathological Complete Response in Early or Locally Advanced Triple-Negative Breast Cancer

7 Jan, 2025 | 12:00h | UTC

Background: Triple-negative breast cancer (TNBC) accounts for approximately 15% of all breast malignancies and is known for its aggressive clinical course and limited therapeutic options. Neoadjuvant chemotherapy has become standard of care for many patients with stage II or III TNBC, aiming to downstage tumors and enhance surgical outcomes. Recent trials suggest that adding immune checkpoint inhibitors, such as anti–programmed death 1/ligand 1 (PD-1/PD-L1) agents, to anthracycline- and platinum-based regimens can further improve response rates. Camrelizumab, a humanized monoclonal anti–PD-1 antibody, has shown antitumor activity in various malignancies, including advanced TNBC. This study, the CamRelief trial, evaluated whether camrelizumab combined with intensive chemotherapy (nab-paclitaxel plus carboplatin followed by dose-dense anthracycline-cyclophosphamide) increases pathological complete response (pCR) in patients with early or locally advanced TNBC.

Objective: To determine if the addition of camrelizumab to a platinum-containing neoadjuvant chemotherapy regimen significantly improves pCR in operable or locally advanced TNBC compared with placebo plus the same chemotherapy backbone.

Methods: This randomized, double-blind, phase 3 trial enrolled 441 female patients aged 18 to 75 years from 40 Chinese centers. Eligible participants had stage II or III TNBC (T2N0-1M0/T3N0M0 or T2N2-3M0/T3N1-3M0) and an Eastern Cooperative Oncology Group performance-status score of 0 or 1. Patients were randomized 1:1 to receive camrelizumab (200 mg) or placebo plus chemotherapy. Chemotherapy consisted of nab-paclitaxel (100 mg/m^2) and carboplatin (area under the curve, 1.5) on days 1, 8, and 15 every 28 days for 16 weeks, then epirubicin (90 mg/m^2) and cyclophosphamide (500 mg/m^2) every two weeks for 8 weeks. Camrelizumab or placebo was administered every two weeks during the 24 weeks of chemotherapy. The primary end point was pCR (ypT0/Tis ypN0), assessed by a local pathologist masked to treatment assignment, at the time of surgery. Secondary end points included event-free survival, disease-free survival, distant disease-free survival, and safety.

Results: Of the 441 randomized patients, 89.2% in the camrelizumab-chemotherapy group and 91.3% in the placebo-chemotherapy group proceeded to surgery. Pathological complete response was observed in 56.8% (95% CI, 50.0%-63.4%) of patients receiving camrelizumab-chemotherapy vs 44.7% (95% CI, 38.0%-51.6%) of those receiving placebo-chemotherapy (rate difference, 12.2% [95% CI, 3.3%-21.2%]; 1-sided P = .004). Serious adverse events occurred more frequently with camrelizumab-chemotherapy (34.7% vs 22.8%), including decreased neutrophil count, decreased platelet count, and decreased white blood cell count, but most immune-related events (eg, reactive capillary endothelial proliferation, hypothyroidism) were low grade and manageable. Longer-term outcomes (event-free survival, disease-free survival, distant disease-free survival) remain immature at a median follow-up of 14.4 months, with hazard ratios of 0.80 (95% CI, 0.46-1.42), 0.58 (95% CI, 0.27-1.24), and 0.62 (95% CI, 0.29-1.33), respectively.

Conclusions: In patients with early or locally advanced TNBC, adding camrelizumab to a platinum-containing, dose-dense anthracycline-cyclophosphamide regimen significantly increased pCR rates compared with placebo plus the same chemotherapy backbone. Safety profiles were consistent with known effects of camrelizumab and dose-dense chemotherapy, with no unexpected toxicities detected.

Implications for Practice: These findings support the integration of camrelizumab into an intensive neoadjuvant regimen for TNBC, potentially offering higher rates of tumor eradication across various nodal stages. Clinicians should, however, anticipate and monitor immune-related adverse events, particularly in higher-risk populations. Given the toxicity profile, patient selection and vigilant supportive care are important.

Study Strengths and Limitations: Strengths include the randomized, double-blind design and the inclusion of higher-risk, node-positive cohorts (including N3). The dose-dense chemotherapy backbone further strengthens the applicability of results in more advanced operable disease. Limitations involve the short follow-up time, which precludes conclusive survival analyses, and the study’s restriction to a single geographic population.

Future Research: Longer follow-up is necessary to determine if improved pCR translates into sustained survival benefits. Biomarker-driven approaches, including further PD-L1 and immunogenomic analyses, may refine patient selection. Studies in broader populations and direct comparisons with other checkpoint inhibitors will help define the optimal immunotherapy-partner regimens in TNBC.

Reference:

Chen L, Li H, Zhang H, et al. Camrelizumab vs Placebo in Combination With Chemotherapy as Neoadjuvant Treatment in Patients With Early or Locally Advanced Triple-Negative Breast Cancer: The CamRelief Randomized Clinical Trial. JAMA. Published online December 13, 2024. DOI: http://doi.org/10.1001/jama.2024.23560

Joensuu H. Neoadjuvant Camrelizumab for Triple-Negative Breast Cancer. Editorial. JAMA. Published online December 13, 2024. DOI: http://doi.org/10.1001/jama.2024.25927


Clinical Overview of Cervical Cancer: Screening, Treatment, and Future Directions

6 Jan, 2025 | 10:00h | UTC

Introduction: This summary provides a concise review of a comprehensive guideline on cervical cancer, covering its epidemiology, risk factors, clinical presentation, and current therapeutic strategies. The aim is to highlight best practices for prevention, screening, and management, as well as emerging treatments that may shift the standard of care.

Key Recommendations:

  1. Prevention and Screening
    • Encourage HPV vaccination before exposure, ideally in adolescence.
    • Perform regular screening with a Papanicolaou test, HPV testing, or both, based on national guidelines.
    • Use colposcopy and directed biopsies for women with abnormal screening results.
  2. Early-Stage Disease (FIGO IA to IB2)
    • Offer radical hysterectomy plus pelvic lymphadenectomy; ovarian preservation may be considered for endocrine benefits.
    • Less radical surgery (simple hysterectomy or conization) is now acceptable for smaller tumors (<2 cm) confirmed by imaging and pathology.
    • In selected cases, fertility-sparing radical trachelectomy can be considered, though the SHAPE trial supports more conservative approaches for certain early tumors.
  3. Locally Advanced Disease (FIGO IB3 to IVA)
    • Recommend concurrent chemoradiation therapy (daily external-beam radiotherapy, brachytherapy, and weekly cisplatin).
    • Immunotherapy (pembrolizumab) is approved in combination with chemoradiation for FIGO III to IVA disease, demonstrating improved survival.
    • Ensure treatment completion within optimal time frames to maximize therapeutic efficacy.
  4. Pelvic Exenteration
    • Consider total pelvic exenteration for isolated central recurrence in patients without distant disease.
    • Thorough psychosocial evaluation is critical before proceeding with this extensive procedure.
  5. Metastatic or Recurrent Disease (First-Line Therapy)
    • A platinum-based regimen (cisplatin or carboplatin) combined with paclitaxel, with or without bevacizumab, remains a standard option.
    • For PD-L1–positive tumors, adding pembrolizumab has shown a survival advantage.
    • Bispecific immunotherapy and novel therapeutic regimens are under investigation to improve outcomes further.
  6. Second-Line Therapy
    • Tissue factor–directed antibody–drug conjugates (e.g., tisotumab vedotin) are effective for patients whose disease progresses after frontline therapy.
    • HER2-targeted therapies (e.g., trastuzumab deruxtecan) may benefit individuals with HER2-overexpressing cervical tumors.
    • Immune checkpoint inhibitors (cemiplimab, nivolumab) can be used in PD-L1–positive recurrent disease, although prior treatment with pembrolizumab may affect efficacy.

Conclusion: By combining targeted vaccination, robust screening programs, and multidisciplinary treatment strategies, cervical cancer can be dramatically reduced worldwide. Advanced management incorporates state-of-the-art surgical procedures, chemoradiation, immunotherapies, and emerging targeted therapies to extend survival and enhance quality of life. Ongoing research aims to optimize treatment sequencing, define new biomarkers, and advance global eradication efforts.

Reference:
Tewari KS. Cervical Cancer. New England Journal of Medicine. (2025). Link: https://www.nejm.org/doi/full/10.1056/NEJMra2404457

 


Systemic Therapy for Stage I-III Anal Squamous Cell Carcinoma: Highlights of the ASCO Guideline

6 Jan, 2025 | 09:00h | UTC

Introduction:
This summary presents the key points of the American Society of Clinical Oncology (ASCO) guideline on systemic therapy for patients with stage I-III anal squamous cell carcinoma (SCC). The guideline’s objectives are to offer evidence-based recommendations that support clinicians in selecting radiosensitizing chemotherapy, dosage regimens, treatment strategies, the use of induction chemotherapy, and the use of ongoing adjuvant chemotherapy. This guidance focuses on optimizing chemoradiation (CRT) to improve oncologic outcomes while minimizing toxicities, particularly the myelosuppression that can limit therapy tolerability. Patients who are immunosuppressed and those with comorbidities receive special consideration.

Key Recommendations:

Radiosensitizing Chemotherapy

  • Mitomycin-C (MMC) + Fluoropyrimidine: The standard radiosensitizing combination is MMC with fluorouracil (FU). Radiosensitizing means making the cancer cells more sensitive to radiation therapy. MMC with capecitabine (an oral alternative to FU) may also be offered, especially when infusion access is a concern. However, MMC is linked to higher hematologic toxicity, so its use demands vigilant monitoring.
  • Cisplatin + FU: An alternative option for radiosensitization. This combination demonstrated noninferiority to MMC + FU in the ACT-II trial. The guideline states that the preferable regimen for patients with immunosuppression is cisplatin and FU, due to the myelosuppression associated with MMC. However, this regimen is not limited to this population. Cisplatin is unsuitable for individuals with renal impairment, significant neuropathy, or hearing loss, and there is no evidence supporting carboplatin substitution.

Dose and Schedule

  • MMC + FU: Common regimens include MMC 10 mg/m^2 on days 1 and 29 (with caution on the second dose) or a single dose of 12 mg/m^2 on day 1, along with continuous-infusion FU (1,000 mg/m^2) on days 1–4 and 29–32. Clinicians should note that there is ongoing discussion about giving one versus two MMC doses, given the additional hematologic toxicity and radiation breaks often observed with two cycles.
  • MMC + Capecitabine: MMC (single or divided dose as above) plus capecitabine (825 mg/m^2 orally twice daily on radiation days) is often used in practice, although large randomized trial data are lacking.
  • Cisplatin + FU: Most commonly, cisplatin 60 mg/m^2 on days 1 and 29, with continuous-infusion FU (1,000 mg/m^2) on days 1–4 and 29–32, can be used. Weekly cisplatin regimens (20 mg/m^2 plus FU 300 mg/m^2) are another acceptable approach, though based on a lower level of evidence.

Single-Agent FU
For patients deemed unable to tolerate combination chemotherapy (e.g., poor performance status), single-agent FU with concurrent radiation may be offered.

Induction and Adjuvant Chemotherapy
No survival or disease-control benefit was observed with adding induction chemotherapy before CRT, nor with additional chemotherapy after CRT for localized anal cancer. Hence, routine use of induction or ongoing adjuvant therapy is not recommended.

Conclusion:
The guideline’s recommendations are based on moderate-quality evidence. These recommendations reinforce the longstanding role of MMC plus FU as the preferred radiosensitizing regimen for stage I-III anal SCC, with cisplatin-based or capecitabine-based options for specific patient needs. The guideline highlights that there are disparities in anal cancer incidence and outcomes, with higher rates among Black men and MSM. These disparities are further complicated by social determinants of health, such as smoking rates, HPV vaccination coverage, and access to screening and treatment. Limiting treatment toxicity—especially myelosuppression—remains critical to preserve treatment adherence and minimize breaks in radiation. Clinicians should tailor therapy to each patient’s comorbidities and performance status. Meanwhile, ongoing trials—such as ECOG-ACRIN 2165 (NCT03233711)—are investigating immunotherapy approaches for higher-risk locally advanced anal cancer, potentially informing future guideline updates.

Reference: Morris VK, Kennedy EB, Amin MA, et al. “Systemic Therapy for Stage I-III Anal Squamous Cell Carcinoma: ASCO Guideline.” Journal of Clinical Oncology. https://doi.org/10.1200/JCO-24-02120

 


RCT: Pembrolizumab with Preoperative Radiotherapy Shows Potential in Stage III Soft Tissue Sarcoma

2 Jan, 2025 | 09:00h | UTC

Background: Patients with locally advanced, high-grade soft tissue sarcomas of the extremity often face a high risk of metastatic disease, despite curative-intent surgery and radiotherapy. Traditional doxorubicin-based chemotherapy provides variable benefits and can cause considerable toxicity, prompting investigations into alternative strategies. Emerging data from smaller trials hinted that immune checkpoint inhibitors might offer targeted benefit in sarcomas, but robust evidence in the neoadjuvant setting remains limited.

Objective: To assess whether adding neoadjuvant and adjuvant pembrolizumab to preoperative radiotherapy and surgical resection could enhance disease-free survival (DFS) in individuals with resectable grade 2 or 3, stage III undifferentiated pleomorphic sarcoma or liposarcoma of the extremity and limb girdle.

Methods: This open-label, randomized trial (SU2C-SARC032) enrolled 143 participants at 20 academic centers in Australia, Canada, Italy, and the USA. Eligible patients were 12 years or older, presented with primary tumors >5 cm, and did not receive chemotherapy as part of this protocol. Participants were randomized 1:1 to standard preoperative radiotherapy (50 Gy/25 fractions) plus surgery (control) or the same radiotherapy combined with pembrolizumab (200 mg every three weeks) in the neoadjuvant setting, followed by up to 14 adjuvant cycles. The primary endpoint was disease-free survival, analyzed in a modified intention-to-treat cohort of 127 evaluable patients, with a median follow-up of 43 months.

Results: The pembrolizumab group demonstrated a higher 2-year DFS rate (67%) compared with controls (52%), suggesting a favorable hazard ratio (0.61) for recurrence or death. Nonetheless, grade 3 or higher adverse events were more common in the pembrolizumab arm (56% vs 31%). Secondary endpoints, including distant disease-free survival and overall survival, also appeared to favor the pembrolizumab arm, but these comparisons were not powered for definitive conclusions.

Conclusions: Neoadjuvant and adjuvant pembrolizumab in combination with radiotherapy and surgery demonstrated a DFS advantage in stage III undifferentiated pleomorphic sarcoma or liposarcoma, reinforcing the potential role of immunotherapy in high-risk settings. However, given the increased incidence of adverse events and the relatively short follow-up for overall survival, cautious interpretation is warranted. Further evidence is required to determine long-term benefits and confirm whether these findings extend to other sarcoma subtypes.

Implications for Practice: These data suggest that incorporating pembrolizumab could be considered in selected patients, particularly those with large, high-grade tumors unresponsive to or unsuitable for traditional chemotherapy. Clinicians must balance the incremental risk of immunotherapy-induced side effects against the observed gains in disease-free survival and the ongoing need for extended follow-up.

Study Strengths and Limitations: Strengths include a multicenter randomized design, focus on a high-risk population, and a robust primary endpoint. Limitations encompass a relatively small sample size, inherent to rare cancers, underpowered subgroup analyses, and absence of long-term survival data. Confirmation of these early signals in larger cohorts and over more extended follow-up periods remains necessary.

Future Research: Additional trials should explore optimal radiotherapy fractionation, potential synergies with cytotoxic or targeted agents, and predictive biomarkers of response. Understanding immune correlates, including circulating tumor DNA and tumor microenvironmental factors, may refine treatment selection and enhance therapeutic outcomes.

Reference: Mowery YM, et al. Safety and efficacy of pembrolizumab, radiation therapy, and surgery versus radiation therapy and surgery for stage III soft tissue sarcoma of the extremity (SU2C-SARC032): an open-label, randomised clinical trial. The Lancet. 2024;404(10467). DOI: http://doi.org/10.1016/S0140-6736(24)01812-9

 


Meta-analysis: One-day Low-residue Diet Achieves Comparable Bowel Cleansing Compared to Multi-day Regimens

26 Dec, 2024 | 18:21h | UTC

Background: Colorectal cancer remains a leading cause of cancer-related morbidity worldwide, making early detection through colonoscopy essential. Adequate bowel preparation is crucial to maximize mucosal visibility and detect lesions effectively. Although low-residue diets (LRDs) are commonly recommended before colonoscopy, guidelines vary regarding the optimal duration (one day versus multiple days). This systematic review and meta-analysis evaluated whether a one-day LRD regimen is non-inferior to multi-day protocols in achieving satisfactory bowel cleansing and lesion detection.

Objective: To compare the efficacy of 1-day versus >1-day LRD regimens for bowel preparation in adult patients undergoing elective colonoscopy, focusing on bowel cleanliness, polyp detection, and adenoma detection rates.

Methods: A comprehensive search of PubMed, Cochrane Central Register of Controlled Trials, ScienceDirect, Scopus, and ClinicalTrials.gov was conducted for randomized controlled trials (RCTs) comparing 1-day with >1-day LRD regimens. Six RCTs involving 2,469 participants met inclusion criteria. Patients were randomized to either a 1-day LRD (n=1,237) or a multi-day LRD (n=1,232). Adequate bowel preparation was primarily defined by a Boston Bowel Preparation Scale (BBPS) score ≥2 in each segment or total BBPS ≥6. Secondary outcomes included polyp detection rate (PDR), adenoma detection rate (ADR), withdrawal time, cecal intubation rate, and cecal intubation time.

Results: Both groups demonstrated similar rates of adequate bowel preparation (87.2% in the 1-day LRD vs. 87.1% in the multi-day group), with no significant difference (OR=1.03, 95% CI, 0.76–1.41; p=0.84; I2=0%). PDR was likewise comparable (OR=0.91, 95% CI, 0.76–1.09; p=0.29; I2=16%), as was ADR (OR=0.87, 95% CI, 0.71–1.08; p=0.21; I2=0%). Withdrawal time did not differ (MD=–0.01 minutes, 95% CI, –0.25 to 0.24; p=0.97; I2=63%), and cecal intubation parameters were also statistically similar. Across studies, the pooled mean global BBPS revealed minimal difference (MD=0.16, 95% CI, –0.02 to 0.34; p=0.08; I2=15%), confirming the non-inferiority of a shorter LRD protocol.

Conclusions: A one-day LRD achieves bowel cleansing outcomes comparable to those of multi-day LRDs, without compromising polyp or adenoma detection. This shorter regimen may help optimize patient adherence, reduce dietary restriction burden, and simplify procedural logistics, especially for busy endoscopy practices.

Implications for Practice: Adopting a 1-day LRD can streamline preparation, improve patient satisfaction, and maintain high-quality visualization. Clinicians should weigh individual patient factors such as chronic constipation or comorbidities but may generally favor a shorter dietary restriction period to enhance compliance and comfort.

Study Strengths and Limitations: This meta-analysis included only RCTs, strengthening its internal validity. Heterogeneity for primary outcomes was minimal. However, the included trials employed varied dietary protocols and bowel preparation solutions. Additionally, some studies lacked uniform reporting of cecal intubation endpoints, limiting direct comparisons. Future investigations with standardized outcome measures could offer more definitive guidance.

Future Research: Further large-scale RCTs should assess cost-effectiveness, patient-reported outcomes, and LRD composition in specific populations. Identifying optimal dietary instructions for individuals with slower colonic transit or specific nutritional needs would refine colonoscopy preparation guidelines and potentially increase detection of precancerous lesions.

Reference: Putri RD, et al. One-day low-residue diet is equally effective as the multiple-day low-residue diet in achieving adequate bowel cleansing: a meta-analysis of randomized controlled trials. Clinical Endoscopy. 2024. DOI: https://doi.org/10.5946/ce.2024.061

 


RCT: Daratumumab Monotherapy Prevents Progression in High-Risk Smoldering Multiple Myeloma

26 Dec, 2024 | 15:44h | UTC

Background: Smoldering multiple myeloma (SMM) is an asymptomatic plasma cell disorder that can progress to active multiple myeloma, especially when risk factors place patients in a high-risk subset. Although daratumumab has been approved for multiple myeloma, no treatments have been approved for high-risk SMM. This study (AQUILA) examined whether subcutaneous daratumumab could prevent or significantly delay progression to symptomatic myeloma.

Objective: To evaluate the effectiveness of subcutaneous daratumumab monotherapy versus active monitoring in prolonging time to disease progression (defined by SLiM–CRAB criteria) or death in patients with high-risk SMM.

Methods: In this open-label phase 3 trial, 390 patients with high-risk SMM were randomly assigned (1:1) to either daratumumab (1800 mg subcutaneously) or active monitoring. Daratumumab was administered weekly for cycles 1–2, every two weeks for cycles 3–6, and then every four weeks for up to 39 cycles (36 months) or until confirmed disease progression. Active monitoring followed the same schedule for disease evaluations without any specific therapy. The primary endpoint was progression-free survival (PFS), assessed by an independent committee. Secondary endpoints included overall survival, response rates, and time to subsequent therapy.

Results: After a median follow-up of 65.2 months, disease progression or death occurred in 34.5% of patients in the daratumumab group compared to 50.5% in the active-monitoring group (HR, 0.49; 95% CI, 0.36–0.67; p<0.001). At five years, the PFS rate was 63.1% with daratumumab and 40.8% with active monitoring. Overall survival was also higher in the daratumumab arm: 93.0% versus 86.9% at five years (HR for death, 0.52; 95% CI, 0.27–0.98). Treatment discontinuation due to adverse events was low (5.7%), and no new safety signals emerged. Grade 3 or 4 adverse events, primarily hypertension (5.7% vs. 4.6%), occurred at similar rates in both arms. Infections of grade 3 or 4 were more frequent with daratumumab (16.1% vs. 4.6%), including COVID-19 pneumonia, yet overall tolerability remained acceptable. Patient-reported outcomes, including quality-of-life measures, were largely preserved in both groups during the study.

Conclusions: Subcutaneous daratumumab monotherapy substantially delayed progression to symptomatic multiple myeloma and improved overall survival among patients with high-risk SMM. The safety profile was consistent with prior daratumumab studies, suggesting a favorable risk–benefit balance. Early intervention with daratumumab may thus alter the disease trajectory for select patients, sparing them from end-organ damage and improving long-term clinical outcomes.

Implications for Practice: While active monitoring has been the standard of care for high-risk SMM, these findings support early therapeutic intervention for patients with multiple high-risk features. Clinicians should remain cautious, however, when generalizing across different risk stratification models. Additional research on optimal treatment durations, combination strategies, and real-world outcomes will further refine patient selection and management of high-risk SMM.

Study Strengths and Limitations: This trial featured robust follow-up (median of over five years) and clear outcome definitions. However, the classification of high-risk features has evolved, and certain populations (e.g., Black patients) were underrepresented. These factors may limit the generalizability of the findings in broader clinical settings.

Future Research: Ongoing trials are investigating alternative dosing schedules, combination regimens (e.g., daratumumab-based quadruplets), and the role of minimal residual disease monitoring to optimize patient outcomes. Additional studies will clarify whether more intense or shorter treatments might maintain efficacy with fewer side effects.

Reference: Dimopoulos MA, Voorhees PM, Schjesvold F, Cohen YC, Hungria V, Sandhu I, Lindsay J, +29, for the AQUILA Investigators. Daratumumab or Active Monitoring for High-Risk Smoldering Multiple Myeloma. New England Journal of Medicine. 2024; DOI: http://doi.org/10.1056/NEJMoa2409029

 


RCT: Avoiding Prophylactic Drain Increases Postoperative Invasive Procedures After Gastrectomy

25 Dec, 2024 | 12:47h | UTC

Background: Prophylactic abdominal drainage following gastrectomy for gastric cancer has been debated for decades. While some Enhanced Recovery After Surgery (ERAS) guidelines discourage routine drains, many surgeons still advocate their use to detect and manage intra-abdominal collections before they become severe. Previous trials were small and underpowered, thus failing to provide robust evidence regarding the real need for prophylactic drains.

Objective: To determine whether omitting a prophylactic drain in gastric cancer surgery leads to a higher likelihood of postoperative invasive procedures (reoperation or percutaneous drainage) within 30 days.

Methods: In this multicenter randomized clinical trial, 404 patients from 11 Italian centers were randomly assigned to either prophylactic drain placement or no drain at the end of subtotal or total gastrectomy. Both academic and community hospitals participated. The primary composite outcome was the rate of reoperation or percutaneous drainage within 30 postoperative days, analyzed via a modified intention-to-treat approach. Secondary endpoints included overall morbidity, anastomotic leaks, length of hospital stay, and 90-day mortality. A parallel invited commentary addressed methodological and clinical perspectives.

Results: Among the 390 patients who underwent resection, 196 had a prophylactic drain and 194 did not. By postoperative day 30, 7.7% of patients in the drain group required reoperation or percutaneous drainage, compared with 15% in the no-drain group. This statistically significant difference was driven by a higher reoperation rate in patients without drains. Both groups had similar anastomotic leak rates (approximately 4% overall). However, patients without prophylactic drains had a higher in-hospital mortality (4.6% vs 0.5%) and were more likely to require escalation of care. There were few drain-related complications, indicating a low risk associated with drain placement. Length of hospital stay and readmission rates were comparable between groups.

Conclusions: Omitting prophylactic drains in gastrectomy was associated with an increased need for postoperative invasive interventions, particularly reoperations. While prior guidelines have recommended against routine drain placement, these findings challenge that stance for total and even subtotal gastrectomies. Surgeons may wish to revisit existing protocols, especially in facilities with fewer resources or lower patient volumes, given the potential reduction in reoperation risk associated with prophylactic drainage.

Implications for Practice: Clinicians should carefully balance possible benefits (earlier detection of fluid collections and reduced reoperations) against potential drawbacks of drain usage. Routine placement may be reconsidered, at least in higher-risk cases or in institutions less equipped for complex salvage procedures.

Study Strengths and Limitations: Key strengths include its robust sample size and standardized criteria for complications. Limitations involve the unblinded nature of postoperative management and the lack of drain fluid amylase measurements to guide removal protocols. Additionally, differentiating total from subtotal gastrectomies might refine selection criteria for prophylactic drainage.

Future Research: Further studies could focus on stratified risk profiles for total vs subtotal gastrectomy and on biomarkers in drain fluid to identify subgroups most likely to benefit from prophylactic drainage.

Reference:
Weindelmayer J, Mengardo V, Ascari F, et al. Prophylactic Drain Placement and Postoperative Invasive Procedures After Gastrectomy: The Abdominal Drain After Gastrectomy (ADIGE) Randomized Clinical Trial. JAMA Surg. Published online November 27, 2024. doi: http://doi.org/10.1001/jamasurg.2024.5227

Invited Commentary: Coffey MR, Lambert KE, Strong VE. Refrain From the Drain? The ADIGE Trial Brings Gastrectomy to the Debate. JAMA Surg. Published online November 27, 2024. doi: http://doi.org/10.1001/jamasurg.2024.5228

 


AGA Clinical Practice Update on Screening and Surveillance in High-Risk US Populations for Gastric Cancer: Expert Review

25 Dec, 2024 | 11:02h | UTC

Introduction:
This American Gastroenterological Association (AGA) Clinical Practice Update provides guidance on primary and secondary prevention strategies for gastric cancer (GC) among high-risk groups in the United States. GC disproportionately affects racial and ethnic minorities, certain first-generation immigrants from countries with elevated GC incidence, and individuals with specific hereditary syndromes or family histories of GC. Given ongoing disparities in diagnosis and outcomes, this document outlines best practices for recognizing at-risk individuals, performing high-quality endoscopic screening, and establishing surveillance protocols for gastric precancerous conditions.

Key Recommendations:

  1. Identify High-Risk Groups: Consider screening among first-generation immigrants from high-incidence regions, people with a family history of GC in a first-degree relative, individuals with hereditary gastrointestinal syndromes, and patients with multiple risk factors (eg, chronic Helicobacter pylori infection, smoking, diets high in salt and processed meats).
  2. Preferred Screening Modality: Upper endoscopy is considered the best method for detecting precancerous lesions (atrophic gastritis and intestinal metaplasia) and early malignancies. It allows direct visualization of the gastric mucosa, systematic biopsy, and accurate histologic staging.
  3. High-Quality Endoscopic Examination: Essential elements include high-definition endoscopes, optimal mucosal cleansing and insufflation, adequate inspection time, systematic photodocumentation, and biopsy protocols (such as the updated Sydney System) to detect and characterize precancerous changes or early cancer.
  4. H. pylori Eradication: Opportunistic screening for H. pylori and its eradication are key adjunctive measures in preventing GC development. Family-based testing—screening adult household members of H. pylori–positive individuals—may further reduce reinfection rates and disease progression.
  5. Systematic Biopsy Protocols: When atrophic gastritis or intestinal metaplasia is suspected, obtain at least five biopsies (antrum/incisura and corpus in separate containers). Any suspicious lesion should be sampled independently.
  6. Recognition of Metaplasia and Dysplasia: Endoscopists should be trained to accurately identify visual patterns associated with gastric intestinal metaplasia (GIM) and dysplasia. Artificial intelligence may hold promise, but current data are insufficient to recommend routine use.
  7. Risk Stratification and Surveillance Intervals: Patients with confirmed GIM or dysplasia, especially those with severe or extensive metaplasia, may require follow-up endoscopy every three years. Individuals with multiple risk factors or severe metaplastic changes could benefit from shorter intervals.
  8. Management of Dysplasia and Early GC: All dysplasia should be reviewed by an expert gastrointestinal pathologist. Visible high-grade dysplasia or early GC lesions generally warrant endoscopic submucosal dissection (ESD) at specialized centers to achieve en bloc, R0 resection and enable accurate pathology.
  9. Post-Resection Surveillance: Individuals with successfully resected dysplasia or early cancer need ongoing endoscopic surveillance to detect metachronous lesions. Surveillance intervals vary depending on pathology results and patient-level factors.
  10. De-Escalation of Screening: Discontinue screening or surveillance when the patient is no longer fit for potential endoscopic or surgical treatment.
  11. Equity and Sustainability: To reduce GC mortality, it is crucial to address modifiable risk factors, enhance patient access to endoscopy and skilled practitioners, and integrate research advances, especially in noninvasive biomarker development and improved endoscopic technologies.

Conclusion:
An effective US-based GC screening and surveillance program requires robust preprocedural identification of high-risk individuals, intraprocedural adherence to quality endoscopy standards, and consistent postprocedural follow-up to ensure equitable access to treatment. By refining these clinical practices and prioritizing research, meaningful reductions in GC incidence and mortality can be achieved, ultimately improving patient outcomes and addressing healthcare disparities.

Reference:
Shah SC, Wang AY, Wallace MB, Hwang JH. AGA Clinical Practice Update on Screening and Surveillance in Individuals at Increased Risk for Gastric Cancer in the United States: Expert Review. Gastroenterology. Published online December 23, 2024.
https://doi.org/10.1053/j.gastro.2024.11.001

 


Network Meta-Analysis: Triplet and Quadruplet Chemotherapy Regimens for Advanced Pancreatic Cancer

22 Dec, 2024 | 17:38h | UTC

Background: Advanced pancreatic ductal adenocarcinoma (PDAC) is associated with poor survival, as most patients present with metastatic or unresectable disease. While gemcitabine monotherapy was the mainstay for decades, subsequent trials found survival gains with multi-agent regimens such as FOLFIRINOX and gemcitabine plus nab-paclitaxel. However, head-to-head data comparing these regimens are limited. This systematic review and Bayesian network meta-analysis aimed to consolidate available evidence on first-line treatment options, focusing on both efficacy and safety outcomes.

Objective: To compare different chemotherapy regimens for unresectable locally advanced or metastatic PDAC, determining which options confer the greatest benefit in progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and toxicity profiles.

Methods: Phase 2–3 randomized controlled trials published since 2000 were searched in PubMed, Cochrane Central, and Embase, and from oncology conference proceedings. Eligible studies enrolled previously untreated patients with advanced PDAC, examining at least one regimen containing gemcitabine, fluorouracil, oxaliplatin, irinotecan, nab-paclitaxel, or liposomal irinotecan. Hazard ratios (HRs) and odds ratios (ORs) were computed within a Bayesian framework, ranking therapies using surface under the cumulative ranking curves. Risk of bias was assessed with Cochrane’s RoB 2 tool; evidence certainty was appraised using GRADE.

Results: Seventy-nine trials (22,168 patients) were included. PFS analysis showed gemcitabine plus nab-paclitaxel alternating FOLFOX (HR 0.32), PAXG (0.35), and NALIRIFOX (0.43) offered the highest benefit versus gemcitabine alone, followed by FOLFIRINOX (0.55) and gemcitabine plus nab-paclitaxel (0.62). For OS, the top three regimens were PAXG (HR 0.40), gemcitabine plus nab-paclitaxel alternating FOLFOX (0.46), and NALIRIFOX (0.56), with FOLFIRINOX (0.66) and gemcitabine plus nab-paclitaxel (0.67) close behind. NALIRIFOX demonstrated relatively lower rates of hematologic toxicity compared with FOLFIRINOX and gemcitabine plus nab-paclitaxel, although diarrhea and neuropathy remained frequent across most multi-drug regimens. Overall, the certainty of evidence was low, largely because of indirect comparisons and high heterogeneity among trials.

Conclusions: Among triplet options, NALIRIFOX and FOLFIRINOX appear most favorable for patients who can tolerate intensified therapy, with gemcitabine plus nab-paclitaxel remaining a viable option for less fit patients. Quadruplet regimens, either administered concurrently or sequentially, show promising efficacy but warrant validation in phase 3 trials.

Implications for Practice: Clinical decision-making should balance potential survival benefits with toxicity, patient performance status, and cost-effectiveness considerations. While more intensive combinations can prolong survival, adverse effects and patient quality of life must be carefully monitored.

Study Strengths and Limitations: This is the largest systematic review and network meta-analysis to date in this setting, synthesizing 79 trials. Nevertheless, the evidence base is limited by phase 2 studies, inconsistent reporting of toxicities, and relatively few direct head-to-head comparisons. Most trials lacked central radiographic review.

Future Research: Prospective phase 3 trials evaluating quadruplet or sequential regimens are needed. Additional biomarker-driven strategies, personalized chemotherapy scheduling, and earlier integration of palliative care may also help enhance survival and preserve quality of life in this population.

Reference: Mastrantoni L, Chiaravalli M, Spring A, et al. Comparison of first-line chemotherapy regimens in unresectable locally advanced or metastatic pancreatic cancer: a systematic review and Bayesian network meta-analysis. The Lancet Oncology. 2024;25(12). DOI: http://doi.org/10.1016/S1470-2045(24)00511-4

 


Noninferiority RCT: Omitting Sentinel-Lymph-Node Biopsy Maintains 5-Year Invasive Disease–Free Survival in Early-Stage Breast Cancer

13 Dec, 2024 | 15:15h | UTC

Background: While axillary surgical staging using sentinel-lymph-node biopsy has been a mainstay in early-stage breast cancer management, its necessity in clinically node-negative patients undergoing breast-conserving therapy has been called into question. With tumor biology increasingly guiding treatment decisions, reducing surgical interventions without compromising survival is a major goal.

Objective: To determine whether omitting sentinel-lymph-node biopsy in patients with clinically node-negative, T1 or T2 (≤5 cm) invasive breast cancer undergoing breast-conserving surgery is noninferior to performing sentinel-lymph-node biopsy in terms of 5-year invasive disease–free survival.

Methods: In this prospective, randomized, noninferiority trial, 5502 eligible patients were randomized in a 1:4 ratio to omission of axillary surgery versus sentinel-lymph-node biopsy. The per-protocol population included 4858 patients. All patients received whole-breast irradiation. Invasive disease–free survival, defined as time to any invasive disease event or death, was the primary endpoint. Noninferiority required a 5-year invasive disease–free survival rate ≥85% and a hazard ratio upper limit <1.271.

Results: After a median follow-up of 73.6 months, the 5-year invasive disease–free survival was 91.9% (95% CI, 89.9–93.5) in the omission group and 91.7% (95% CI, 90.8–92.6) in the sentinel-biopsy group (HR, 0.91; 95% CI, 0.73–1.14), meeting noninferiority criteria. Axillary recurrences were slightly higher in the omission group (1.0% vs. 0.3%), though without detrimental effects on overall survival. Patients who omitted axillary surgery experienced lower rates of lymphedema, better arm mobility, and less pain than those undergoing sentinel-lymph-node biopsy.

Conclusions: For appropriately selected patients with clinically node-negative, early-stage breast cancer undergoing breast-conserving surgery, omitting sentinel-lymph-node biopsy did not compromise 5-year invasive disease–free survival and yielded fewer surgical complications and better quality of life outcomes.

Implications for Practice: Omitting axillary staging may be considered in low-risk patients, particularly older individuals with small, hormone receptor-positive, HER2-negative tumors. However, clinicians should balance the lack of nodal information against potential alterations in adjuvant therapy decisions, especially regarding radiotherapy and systemic treatment recommendations.

Study Strengths and Limitations: Strengths include a large patient population, rigorous prospective randomization, and substantial follow-up. Limitations include a predominance of low-risk, small, HR-positive/HER2-negative tumors, potentially limiting generalizability. While omitting sentinel-lymph-node biopsy reduces surgical morbidity, the absence of nodal status may influence adjuvant treatment planning.

Future Research: Further studies should assess the applicability of omission strategies to younger patients, larger tumors, or more aggressive subtypes, and explore whether novel biomarkers or imaging methods can reliably guide treatment decisions without axillary surgery.

Reference: Reimer T, Stachs A, Veselinovic K, Kühn T, Heil J, Polata S, Marmé F, et al. Axillary Surgery in Breast Cancer — Primary Results of the INSEMA Trial. New England Journal of Medicine. 2024. DOI: http://doi.org/10.1056/NEJMoa2412063

 


Nonrandomized Phase 1b–2 Study: CAR T-cell Therapy Obecabtagene Autoleucel Effective with Low Severe Toxicity in Adult B-cell ALL

4 Dec, 2024 | 12:04h | UTC

Background: Relapsed or refractory B-cell acute lymphoblastic leukemia (ALL) in adults has a poor prognosis, with current therapies often requiring allogeneic stem-cell transplantation for durable responses. CAR T-cell therapies targeting CD19 have shown promise but are associated with significant toxic effects.

Objective: To evaluate the efficacy and safety of obecabtagene autoleucel (obe-cel), a novel autologous 41BB-ζ anti-CD19 CAR T-cell therapy designed to reduce toxic effects and improve persistence, in adults with relapsed or refractory B-cell ALL.

Methods: In this nonrandomized, phase 1b–2 multicenter study, 127 adults aged 18 years or older with relapsed or refractory CD19-positive B-cell ALL received at least one infusion of obe-cel. The primary endpoint was overall remission (complete remission or complete remission with incomplete hematologic recovery) in cohort 2A (patients with morphologic disease). Secondary endpoints included event-free survival, overall survival, and safety assessments.

Results: Among the 94 patients in cohort 2A (median follow-up of 20.3 months), overall remission occurred in 77% (95% CI, 67–85%), with 55% achieving complete remission and 21% achieving complete remission with incomplete hematologic recovery. The prespecified null hypotheses were rejected (P < 0.001). In the total infused population, the median event-free survival was 11.9 months (95% CI, 8.0–22.1), and the median overall survival was 15.6 months (95% CI, 12.9–NE). Grade 3 or higher cytokine release syndrome occurred in 2.4% of patients, and grade 3 or higher immune effector cell–associated neurotoxicity syndrome (ICANS) occurred in 7.1%.

Conclusions: Obe-cel demonstrated a high incidence of durable responses with a low incidence of severe immune-related toxic effects in adults with relapsed or refractory B-cell ALL.

Implications for Practice: Obe-cel may offer an effective CAR T-cell therapy option with manageable toxicity for adult patients with relapsed or refractory B-cell ALL. However, long-term benefits and direct comparisons with existing therapies require further investigation. Clinicians should consider the potential advantages but remain cautious given the lack of randomized controlled data.

Study Strengths and Limitations: Strengths include the multicenter design and substantial patient population. Limitations involve the nonrandomized, single-arm design without a control group, and potential bias from patients who did not receive the infusion due to disease progression or manufacturing failures. Additionally, longer follow-up is needed to fully assess durability and late-onset toxicities.

Future Research: Further studies are warranted to compare obe-cel directly with standard therapies in randomized controlled trials, assess long-term outcomes, and explore its efficacy in earlier lines of treatment or in combination with other modalities.

Reference: Roddie C, Sandhu KS, Tholouli E, Logan AC, Shaughnessy P, Barba P, Ghobadi A, et al. Obecabtagene Autoleucel in Adults with B-Cell Acute Lymphoblastic Leukemia. New England Journal of Medicine. Published November 27, 2024. DOI: http://doi.org/10.1056/NEJMoa2406526

 


RCT: Nivolumab Plus Ipilimumab Extends Progression-Free Survival in MSI-H or dMMR Metastatic Colorectal Cancer

4 Dec, 2024 | 11:51h | UTC

Background: Patients with microsatellite-instability–high (MSI-H) or mismatch-repair–deficient (dMMR) metastatic colorectal cancer typically experience poor outcomes with standard chemotherapy. Previous nonrandomized studies suggested that combining nivolumab with ipilimumab may offer clinical benefits in this population.

Objective: To evaluate the efficacy and safety of nivolumab plus ipilimumab compared with chemotherapy in patients with MSI-H or dMMR metastatic colorectal cancer who had not received prior systemic treatment for metastatic disease.

Methods: In this phase 3, open-label, randomized trial, 303 patients with unresectable or metastatic MSI-H or dMMR colorectal cancer were assigned in a 2:2:1 ratio to receive nivolumab plus ipilimumab, nivolumab alone, or chemotherapy with or without targeted therapies. The primary endpoint assessed in this interim analysis was progression-free survival (PFS) of nivolumab plus ipilimumab versus chemotherapy in patients with centrally confirmed MSI-H or dMMR status.

Results: At a median follow-up of 31.5 months, nivolumab plus ipilimumab significantly improved PFS compared to chemotherapy (P<0.001). The 24-month PFS was 72% (95% CI, 64–79) with nivolumab plus ipilimumab versus 14% (95% CI, 6–25) with chemotherapy. The restricted mean survival time at 24 months was 10.6 months longer with the combination therapy. Grade 3 or 4 treatment-related adverse events occurred in 23% of patients receiving nivolumab plus ipilimumab and 48% of those receiving chemotherapy.

Conclusions: First-line treatment with nivolumab plus ipilimumab significantly prolonged progression-free survival compared to chemotherapy in patients with MSI-H or dMMR metastatic colorectal cancer, with a lower incidence of high-grade treatment-related adverse events.

Implications for Practice: The combination of nivolumab and ipilimumab may represent a new standard of care for first-line treatment in MSI-H or dMMR metastatic colorectal cancer. However, clinicians should weigh the benefits against potential immune-related adverse events, and long-term survival benefits remain to be fully established.

Study Strengths and Limitations: Strengths include the randomized, phase 3 design and central confirmation of MSI-H or dMMR status. Limitations involve the open-label design, potential bias in patient-reported outcomes, underrepresentation of certain populations, and immature overall survival data.

Future Research: Further studies are needed to compare nivolumab plus ipilimumab directly with nivolumab monotherapy and to assess long-term overall survival benefits and quality of life in diverse patient populations.

Reference: Andre T, et al. Nivolumab plus Ipilimumab in Microsatellite-Instability–High Metastatic Colorectal Cancer. New England Journal of Medicine. 2024;391(21):2014–2026. DOI: http://doi.org/10.1056/NEJMoa2402141

 


News Release: Sacubitril/Valsartan May Reduce Chemotherapy-Induced Cardiotoxicity in High-Risk Cancer Patients

20 Nov, 2024 | 18:38h | UTC

Introduction: A recent study presented at the American Heart Association’s Scientific Sessions 2024 introduces sacubitril/valsartan, a widely used heart failure medication, as a potential protective agent against heart damage in high-risk cancer patients undergoing anthracycline chemotherapy. Anthracyclines, while effective for treating various cancers such as breast cancer, leukemia, and lymphoma, carry a significant risk of cardiotoxicity, leading to cardiomyopathy and heart failure. The SARAH trial aimed to evaluate whether sacubitril/valsartan could mitigate this risk and preserve cardiac function during chemotherapy.

Highlights: The SARAH trial was a randomized, double-blind, placebo-controlled study involving 114 high-risk cancer patients at Erasto Gaertner Hospital in Curitiba, Brazil. High risk was defined by elevated high-sensitivity troponin I levels post-anthracycline infusion, indicating early signs of cardiac injury. Participants, predominantly women with breast cancer (81%), were randomized to receive sacubitril/valsartan or a placebo over 24 weeks.

Key findings include:

  • Significant Reduction in Cardiotoxicity: Sacubitril/valsartan was associated with a 77% reduction in the relative risk of cardiotoxicity compared to placebo. Cardiotoxicity was measured as a ≥15% reduction in global longitudinal strain (GLS), a sensitive marker of left ventricular function.
  • Improvement in Cardiac Function: Patients in the sacubitril/valsartan group experienced an average improvement in GLS by 2.55%, whereas those in the placebo group showed an average decline of 6.65%.
  • Dose Titration and Tolerance: The medication was initiated at a dose of 24/26 mg twice daily and titrated every two weeks to a target of 97/103 mg twice daily or the highest tolerated dose without side effects. It was generally well tolerated, with hypotension being more common in the treatment group (14% vs. 1.8%) but no significant differences in other adverse events.
  • Independent of Other Factors: The protective effect of sacubitril/valsartan was consistent regardless of cumulative anthracycline dose, HER2 status, presence of hypertension, or patient age.

Conclusion: The findings from the SARAH trial suggest that sacubitril/valsartan may offer a promising strategy to prevent chemotherapy-induced cardiotoxicity in high-risk cancer patients. By preserving cardiac function during anthracycline treatment, this medication has the potential to improve patient outcomes, enhance quality of life, and reduce the long-term burden of heart failure among cancer survivors. Further research with larger, more diverse populations and extended follow-up is warranted to confirm these results and assess the impact on long-term clinical outcomes.

Source: This research was conducted by the Heart Institute at the University of São Paulo, Brazil, and presented at the American Heart Association Scientific Sessions 2024.

Primary Source: American Heart Association News Release
https://newsroom.heart.org/news/a-common-heart-failure-medication-may-help-prevent-heart-damage-related-to-chemotherapy-6906417

Secondary Sources:

 


Cohort Study: Levonorgestrel IUD Use Linked to Increased Breast Cancer Risk in Premenopausal Women

20 Oct, 2024 | 18:13h | UTC

Background: Levonorgestrel-releasing intrauterine systems (LNG-IUSs) are increasingly used, especially among Danish premenopausal women over 30 years old, as a preferred method of hormonal contraception. Previous studies have suggested an increased risk of breast cancer with LNG-IUS use but did not adequately address the duration of continuous use or account for other hormonal contraceptive exposures.

Objective: To assess the risk of breast cancer associated with continuous use of LNG-IUSs, accounting for other hormonal exposures.

Methods: In this nationwide Danish cohort study, 78,595 first-time LNG-IUS users aged 15–49 years from 2000 to 2019 were identified and matched 1:1 by birth year to nonusers of hormonal contraceptives. Exclusion criteria included prior hormonal contraceptive use within 5 years, previous cancer, postmenopausal hormone therapy, and pregnancy at baseline. Participants were followed from initiation until breast cancer diagnosis, other cancer, pregnancy, hormone therapy initiation, emigration, death, or December 31, 2022. Cox proportional hazards models adjusted for confounders estimated hazard ratios (HRs) for breast cancer associated with continuous LNG-IUS use.

Results: During a mean follow-up of 6.8 years, 1,617 breast cancer cases occurred: 720 among LNG-IUS users and 897 among nonusers. The mean age was 38 years. Continuous LNG-IUS use was associated with a higher breast cancer risk compared to nonuse (HR, 1.4; 95% CI, 1.2–1.5). HRs by duration were 1.3 (95% CI, 1.1–1.5) for 0–5 years, 1.4 (95% CI, 1.1–1.7) for >5–10 years, and 1.8 (95% CI, 1.2–2.6) for >10–15 years. Excess breast cancer cases per 10,000 users were 14 (95% CI, 6–23), 29 (95% CI, 9–50), and 71 (95% CI, 15–127), respectively. The trend test for duration was not statistically significant (P = .15).

Conclusions: Continuous use of LNG-IUSs was associated with an increased risk of breast cancer among women aged 15–49 years compared to nonuse of hormonal contraceptives. The absolute increase in risk was low.

Implications for Practice: Healthcare providers should inform women about the potential increased breast cancer risk associated with LNG-IUS use, especially considering its widespread and long-term use among premenopausal women. While the absolute risk increase is small, this information is essential for making informed contraceptive choices.

Study Strengths and Limitations: Strengths include the large, nationwide cohort and adjustment for multiple confounders. Limitations include potential underestimation of risk due to unrecorded LNG-IUS removals before the recommended duration, lack of a statistically significant trend with duration suggesting possible low statistical precision or non-causal association, and the possibility of unmeasured confounding.

Future Research: Further studies are needed to confirm these findings, clarify the causal relationship, and understand the mechanisms underlying the potential increased breast cancer risk with LNG-IUS use.

Reference: Mørch LS, Meaidi A, Corn G, et al. Breast Cancer in Users of Levonorgestrel-Releasing Intrauterine Systems. JAMA. Published online October 16, 2024. DOI: http://doi.org/10.1001/jama.2024.18575

 


Psychedelic-Assisted Therapy May Reduce Anxiety and Depression in Patients with Life-Threatening Diseases

20 Oct, 2024 | 18:02h | UTC

Background: Anxiety, depression, and existential distress are prevalent among individuals facing life-threatening illnesses, significantly impacting their quality of life. Traditional treatments often have limited efficacy in this population. Psychedelic-assisted therapy, involving substances like psilocybin and LSD under professional supervision, has been proposed as a potential intervention. However, these substances are illegal in most countries and pose potential risks.

Objective: To assess the benefits and harms of psychedelic-assisted therapy compared to placebo or active comparators in treating anxiety, depression, and existential distress in people with life-threatening diseases.

Methods: This Cochrane systematic review included six randomized controlled trials conducted in the USA and Switzerland between 2011 and 2022. A total of 149 participants (140 analyzed), aged 36 to 64 years with life-threatening illnesses (e.g., cancer), were randomized to receive psychedelic-assisted therapy using classical psychedelics (psilocybin or LSD) or MDMA. Interventions included preparatory sessions, the psychedelic experience, and integration sessions. Comparators were active placebos (e.g., low-dose psychedelic or niacin) or placebo. Primary outcomes were anxiety, depression, and existential distress measured 1 to 12 weeks post-intervention.

Results: Psychedelic-assisted therapy with classical psychedelics may reduce anxiety and depression compared to active placebo:

  • Anxiety: Mean difference (MD) of −8.41 points on the STAI-Trait scale (20–80 range; 95% CI, −12.92 to −3.89; 5 studies, 122 participants; low-certainty evidence).
  • Depression: MD of −4.92 points on the Beck Depression Inventory (0–63 range; 95% CI, −8.97 to −0.87; 4 studies, 112 participants; low-certainty evidence).

The effect on existential distress was mixed and very uncertain. No treatment-related serious adverse events or grade 3/4 adverse events were reported. Common mild to moderate adverse events included elevated blood pressure, nausea, anxiety, and transient psychotic-like symptoms, which resolved shortly after the sessions.

Conclusions: Psychedelic-assisted therapy with classical psychedelics may reduce symptoms of anxiety and depression in patients with life-threatening diseases, but the evidence is of low certainty due to methodological limitations and small sample sizes. The effects of MDMA-assisted therapy are very uncertain.

Implications for Practice: While findings are promising, clinicians should exercise caution due to the low certainty of evidence and legal restrictions surrounding psychedelic substances.

Study Strengths and Limitations: Strengths include randomized designs and standardized therapeutic protocols involving preparation and integration sessions. Limitations are high risk of bias due to unblinding, small sample sizes, potential expectation bias, and cross-over designs with carry-over effects.

Future Research: Larger, well-designed RCTs with rigorous blinding are needed to confirm these findings. Future studies should explore long-term outcomes, diverse patient populations, and strategies to mitigate bias, such as using active placebos and measuring expectancy effects.

Reference: Schipper S, et al. (2024). Psychedelic-assisted therapy for treating anxiety, depression, and existential distress in people with life-threatening diseases. Cochrane Database of Systematic Reviews. DOI: https://doi.org/10.1002/14651858.CD015383.pub2

 


RCT: Five-Fraction SBRT Noninferior to Conventional Radiotherapy in Localized Prostate Cancer

20 Oct, 2024 | 15:46h | UTC

Background: Prostate cancer poses a significant global health challenge, with radiotherapy being a common curative treatment for localized disease. Hypofractionation, delivering higher doses per session over fewer treatments, has potential benefits in efficacy and convenience. While moderately hypofractionated radiotherapy is established, the efficacy of stereotactic body radiotherapy (SBRT) delivering radiation in just five fractions remains uncertain.

Objective: To assess whether five-fraction SBRT is noninferior to conventionally or moderately hypofractionated radiotherapy regarding freedom from biochemical or clinical failure in patients with low-to-intermediate-risk localized prostate cancer.

Methods: In this phase 3, international, open-label randomized controlled trial (PACE-B), 874 men with stage T1–T2 prostate cancer, Gleason score ≤3+4, and prostate-specific antigen (PSA) ≤20 ng/mL were randomized 1:1 to receive SBRT (36.25 Gy in 5 fractions over 1–2 weeks) or control radiotherapy (78 Gy in 39 fractions over 7.5 weeks or 62 Gy in 20 fractions over 4 weeks). Androgen-deprivation therapy was not permitted. The primary endpoint was freedom from biochemical or clinical failure.

Results: Between August 2012 and January 2018, 874 patients were randomized (433 to SBRT and 441 to control radiotherapy) at 38 centers. Median age was 69.8 years, median PSA was 8.0 ng/mL, and 91.6% had intermediate-risk disease. At a median follow-up of 74.0 months, the 5-year incidence of freedom from biochemical or clinical failure was 95.8% in the SBRT group and 94.6% in the control group (unadjusted HR 0.73; 90% CI, 0.48 to 1.12; P=0.004 for noninferiority). Cumulative incidence of late Radiation Therapy Oncology Group (RTOG) grade 2 or higher genitourinary toxic effects at 5 years was higher with SBRT (26.9% vs. 18.3%; P<0.001), while gastrointestinal toxic effects were similar between groups (10.7% vs. 10.2%; P=0.94). Overall survival did not differ significantly (HR for death, 1.41; 95% CI, 0.90 to 2.20).

Conclusions: Five-fraction SBRT was noninferior to conventional or moderately hypofractionated radiotherapy in terms of biochemical or clinical failure in patients with low-to-intermediate-risk localized prostate cancer. SBRT may be an effective treatment option but is associated with a higher incidence of medium-term genitourinary toxic effects.

Implications for Practice: SBRT offers equivalent oncologic efficacy with the convenience of fewer treatment sessions, potentially reducing patient burden and healthcare resource utilization. Clinicians should consider SBRT for eligible patients but must inform them about the increased medium-term risk of genitourinary toxic effects.

Study Strengths and Limitations: Strengths include a large sample size, multicenter design, standardized radiotherapy protocols, and exclusion of hormonal therapy, minimizing confounding factors. Limitations involve the applicability of findings only to patients similar to those in the trial; some may now opt for active surveillance, and results may not extend to higher-risk populations.

Future Research: Further studies are needed to evaluate long-term outcomes of SBRT, its role in higher-risk patients, and strategies to mitigate genitourinary toxic effects.

Reference: van As N., Griffin C., Tree A., et al. (2024). Phase 3 Trial of Stereotactic Body Radiotherapy in Localized Prostate Cancer. New England Journal of Medicine. DOI: http://doi.org/10.1056/NEJMoa2403365

 


RCT: H. pylori Screening Added to Fecal Immunochemical Testing Did Not Reduce Gastric Cancer Incidence or Mortality

4 Oct, 2024 | 11:00h | UTC

Background: Gastric cancer is a leading cause of cancer-related mortality worldwide, particularly in East Asia. Helicobacter pylori infection is a well-established risk factor for gastric cancer development. While eradication therapy may prevent gastric cancer, the effectiveness of community-based H. pylori screening on gastric cancer incidence and mortality remains uncertain.

Objective: To determine whether adding H. pylori stool antigen (HPSA) testing to fecal immunochemical test (FIT) screening reduces gastric cancer incidence and mortality compared to FIT screening alone.

Methods: In a pragmatic randomized clinical trial conducted in Changhua County, Taiwan (2014–2018), 152,503 residents aged 50 to 69 years eligible for biennial FIT screening were randomized to receive an invitation for HPSA testing plus FIT (n = 63,508) or FIT alone (n = 88,995). Participants in the HPSA + FIT group with positive HPSA results were offered antibiotic eradication therapy. Primary outcomes were gastric cancer incidence and mortality, assessed via national cancer and death registries.

Results: Participation rates were higher in the HPSA + FIT group (49.6%) than in the FIT-alone group (35.7%). In the HPSA + FIT group, 38.5% tested positive for HPSA, and 71.4% of these received antibiotic treatment, achieving a 91.9% eradication rate. Over a median follow-up of approximately 5 years, gastric cancer incidence did not differ significantly between the HPSA + FIT and FIT-alone groups (0.032% vs 0.037%; mean difference –0.005%; 95% CI, –0.013% to 0.003%; P = .23). Gastric cancer mortality rates were also similar (0.015% vs 0.013%; mean difference 0.002%; 95% CI, –0.004% to 0.007%; P = .57). Adjusted analyses accounting for participation rates, follow-up duration, and baseline characteristics showed a lower gastric cancer incidence in the HPSA + FIT group (RR 0.79; 95% CI, 0.63–0.98; P = .04), but no difference in mortality (RR 1.02; 95% CI, 0.73–1.40; P = .91). Adverse effects from antibiotics were mild, with abdominal pain or diarrhea occurring in 2.1%.

Conclusions: An invitation to HPSA testing combined with FIT did not significantly reduce gastric cancer incidence or mortality compared to FIT alone over a median follow-up of about 5 years. Adjusted analyses suggest a potential reduction in gastric cancer incidence but not mortality when accounting for participation rates and follow-up duration.

Implications for Practice: Adding H. pylori screening to existing FIT programs may not significantly reduce gastric cancer incidence or mortality in the short term, possibly due to low participation rates, incomplete eradication, and limited follow-up. Clinicians should consider these factors when implementing community-based H. pylori screening and weigh the benefits against resource utilization and patient adherence.

Study Strengths and Limitations: Strengths include a large sample size and integration of HPSA testing into an existing FIT screening infrastructure. Limitations encompass differences in participation rates and baseline characteristics between groups, a relatively short follow-up period, and only 71.4% of HPSA-positive participants receiving eradication therapy, which may have reduced the ability to detect significant effects.

Future Research: Longer-term studies with higher participation and eradication rates are needed to assess the long-term benefits of H. pylori screening on gastric cancer incidence and mortality. Research should explore strategies to improve screening uptake and treatment adherence.

Reference: Lee Y-C, et al. (2024) Screening for Helicobacter pylori to Prevent Gastric Cancer: A Pragmatic Randomized Clinical Trial. JAMA. DOI: http://doi.org/10.1001/jama.2024.14887

 


RCT: Telehealth-Delivered Early Palliative Care Equivalent to In-Person Care in Advanced Lung Cancer

26 Sep, 2024 | 15:06h | UTC

Background: Patients with advanced lung cancer often face a high symptom burden and decreased quality of life (QOL), but access to early palliative care, which can improve these outcomes, remains limited. While telehealth has become increasingly utilized due to the COVID-19 pandemic, it is unclear whether virtual palliative care is as effective as in-person care.

Objective: To compare the effect of early palliative care delivered via secure video vs in-person visits on the quality of life of patients with advanced non–small cell lung cancer (NSCLC).

Methods: This multisite, randomized comparative effectiveness trial enrolled 1250 adults with advanced NSCLC from 22 cancer centers in the US between June 2018 and May 2023. Participants were randomized to receive either early palliative care via video visits or in person every four weeks. The primary outcome was QOL measured by the Functional Assessment of Cancer Therapy-Lung (FACT-L) questionnaire at 24 weeks. Secondary outcomes included caregiver participation in palliative care visits and patient and caregiver satisfaction with care, mood symptoms, coping, and prognostic understanding.

Results: By week 24, patients in both groups reported equivalent QOL scores, with the video visit group scoring a mean of 99.7 compared to 97.7 in the in-person group (difference of 2.0 points, 90% CI, 0.1-3.9; P = .04 for equivalence). Both groups experienced similar improvements in QOL from baseline (mean increase of 8.4 points for video visits and 6.9 points for in-person care). Caregiver participation in palliative care visits was lower in the video visit group (36.6% vs 49.7%; P < .001). No significant differences were found between the groups in caregiver QOL, patient or caregiver satisfaction with care, mood symptoms, or coping strategies.

Conclusions: Early palliative care delivered via telehealth was equivalent to in-person visits in improving QOL for patients with advanced NSCLC. This underscores the potential of telehealth to increase access to essential palliative care services for this population without compromising care quality.

Implications for Practice: Telehealth can provide a feasible alternative to in-person palliative care, especially for patients with advanced lung cancer who face barriers to in-person visits, such as transportation challenges. However, strategies to enhance caregiver involvement in virtual visits may need to be developed.

Study Strengths and Limitations: Strengths include the large, multisite randomized design and the use of validated outcome measures. Limitations involve the COVID-19 pandemic’s impact, which caused some intervention contamination due to unavoidable video visits in the in-person group. Additionally, caregiver participation was lower than expected, potentially limiting the generalizability of results regarding caregiver outcomes.

Future Research: Further studies should explore the long-term impact of telehealth on palliative care outcomes and investigate ways to enhance caregiver involvement in virtual care.

Reference: Greer, J. A., et al. (2024). Telehealth vs In-Person Early Palliative Care for Patients With Advanced Lung Cancer: A Multisite Randomized Clinical Trial. JAMA. DOI: http://doi.org/10.1001/jama.2024.13964

 


RCT: MRI-Guided Biopsy Reduces Overdiagnosis of Clinically Insignificant Prostate Cancer

26 Sep, 2024 | 12:22h | UTC

Background: Overdiagnosis of clinically insignificant prostate cancer is a significant issue in population-based screening programs, primarily when prostate-specific antigen (PSA) testing is followed by systematic biopsy. Magnetic resonance imaging (MRI)-guided biopsies, which avoid systematic biopsies in men with negative MRI results, have shown potential in reducing unnecessary cancer diagnoses. However, long-term data are needed to confirm the safety and efficacy of this approach.

Objective: To evaluate whether MRI-targeted biopsies, when combined with PSA screening, can reduce the detection of clinically insignificant prostate cancer without compromising the identification of clinically significant or advanced disease.

Methods: This population-based, randomized trial in Sweden (GÖTEBORG-2) enrolled 13,153 men aged 50-60 years who underwent PSA screening. Men with PSA levels ≥3 ng/mL were randomized into two groups: (1) MRI-targeted biopsy only in cases with suspicious lesions, or (2) systematic biopsy in all cases with PSA elevation. Screening occurred every 2, 4, or 8 years depending on PSA levels, with follow-up for up to four years. The primary outcome was the detection of clinically insignificant prostate cancer, and secondary outcomes included clinically significant and advanced or high-risk prostate cancer.

Results: After a median follow-up of 3.9 years, the detection of clinically insignificant prostate cancer was significantly lower in the MRI-targeted biopsy group (2.8%) compared to the systematic biopsy group (4.5%), with a relative risk (RR) of 0.43 (95% CI, 0.32-0.57; P < 0.001). The relative risk of detecting clinically significant cancer was 0.84 (95% CI, 0.66-1.07), indicating no significant difference between the two groups. Advanced or high-risk cancers were detected in 15 men in the MRI group and 23 men in the systematic group (RR, 0.65; 95% CI, 0.34-1.24). Severe adverse events occurred in five patients (three in the systematic biopsy group, two in the MRI-targeted biopsy group).

Conclusions: Omitting biopsies in men with negative MRI results substantially reduced the diagnosis of clinically insignificant prostate cancer without increasing the risk of missing clinically significant or advanced cancers. MRI-targeted biopsy strategies can effectively limit overdiagnosis while maintaining safety in screening programs.

Implications for Practice: MRI-targeted biopsies offer a promising strategy to reduce unnecessary cancer diagnoses and avoid overtreatment in prostate cancer screening. Clinicians should consider integrating MRI into prostate cancer screening algorithms, especially in cases with elevated PSA but no MRI-detected lesions. This approach may also decrease biopsy-related complications and patient anxiety.

Study Strengths and Limitations: Strengths of this trial include its population-based design, large sample size, and thorough follow-up. Limitations include its single-center setting in Sweden, which may limit generalizability to more diverse populations, and a modest participation rate of 50%.

Future Research: Further studies should assess the cost-effectiveness of widespread MRI use in prostate cancer screening and explore its utility in diverse populations. Investigations into novel biomarkers that could further refine patient selection for MRI-targeted biopsy are also warranted.

Reference: Hugosson J., et al. (2024). Results after Four Years of Screening for Prostate Cancer with PSA and MRI. N Engl J Med. DOI: https://doi.org/10.1056/NEJMoa2406050

 


Phase 2 RCT: Ponsegromab Shows Promise for the Treatment of Cancer Cachexia

23 Sep, 2024 | 21:48h | UTC

Background: Cancer cachexia is a multifactorial syndrome characterized by weight loss, muscle wasting, and reduced quality of life. Elevated levels of growth differentiation factor 15 (GDF-15), a cytokine, have been associated with cachexia. Ponsegromab, a monoclonal antibody that inhibits GDF-15, has shown potential in reversing cachexia in early studies by improving weight, appetite, and physical activity. This phase 2, randomized, double-blind trial aimed to assess the efficacy and safety of ponsegromab in patients with cancer cachexia and elevated GDF-15 levels.

Objective: To evaluate the impact of ponsegromab on body weight, cachexia symptoms, appetite, physical activity, and safety in cancer cachexia patients with elevated GDF-15 levels.

Methods: In this 12-week study, 187 patients with non-small-cell lung cancer, pancreatic cancer, or colorectal cancer and elevated GDF-15 levels (≥1500 pg/mL) were randomized into four groups: ponsegromab 100 mg, 200 mg, 400 mg, or placebo, administered subcutaneously every four weeks. The primary endpoint was the change in body weight from baseline to week 12. Secondary outcomes included appetite and cachexia symptoms, physical activity measured via digital devices, and safety. The trial also included exploratory endpoints like changes in skeletal muscle mass.

Results:
At 12 weeks, patients treated with ponsegromab showed significant weight gain compared to placebo. The median weight gain differences were 1.22 kg in the 100-mg group, 1.92 kg in the 200-mg group, and 2.81 kg in the 400-mg group. Significant improvements in appetite and cachexia symptoms were observed in the 400-mg group compared to placebo. Physical activity, measured by nonsedentary time, also increased by 72 minutes per day in the 400-mg group. Adverse events were reported by 70% of ponsegromab patients and 80% of placebo patients. Serious adverse events occurred at similar rates across groups, but none were deemed related to treatment. No significant safety concerns were identified.

Conclusions: Ponsegromab effectively increased body weight and improved cachexia symptoms in patients with cancer cachexia and elevated GDF-15 levels, supporting GDF-15’s role as a key driver of cachexia. The findings suggest that ponsegromab may be a promising therapeutic option for managing cancer cachexia, with a favorable safety profile.

Implications for Practice: Ponsegromab could represent a targeted therapy for cancer cachexia, addressing weight loss, appetite, and physical function. Clinicians may consider its use for patients with advanced cancers experiencing cachexia, particularly those with elevated GDF-15 levels.

Study Strengths and Limitations: Strengths of the study include its randomized, double-blind design and the use of objective measures such as digital physical activity tracking. Limitations include the relatively short trial duration, and missing physical activity data for some patients. Additionally, the efficacy of ponsegromab across different baseline levels of GDF-15 requires further investigation.

Future Research: Larger and longer-term trials are necessary to confirm the therapeutic benefit of ponsegromab in cancer cachexia. Future research should explore its impact on survival and assess whether GDF-15 inhibition benefits other conditions associated with elevated GDF-15, such as heart failure and chronic kidney disease.

Reference: Groarke, J. D., et al. (2024). Ponsegromab for the Treatment of Cancer Cachexia. New England Journal of Medicine. DOI: https://doi.org/10.1056/NEJMoa2409515

 


Phase 2 RCT: Axatilimab Demonstrates Efficacy in Refractory Chronic GVHD by Targeting CSF1R-Dependent Macrophages

19 Sep, 2024 | 16:05h | UTC

Background: Chronic graft-versus-host disease (GVHD) is a significant long-term complication of allogeneic hematopoietic stem-cell transplantation, affecting approximately half of recipients and leading to substantial morbidity and mortality. Standard therapies often fail to induce durable responses in patients with refractory or recurrent disease. CSF1R-dependent monocytes and macrophages are key mediators of chronic GVHD, contributing to inflammation and fibrosis. Axatilimab, a CSF1R-blocking antibody, has shown promising activity in early studies.

Objective: To evaluate the efficacy and safety of axatilimab at three different doses in patients with recurrent or refractory chronic GVHD.

Methods: In this phase 2, multinational, randomized study (AGAVE-201), 241 patients aged ≥2 years with active chronic GVHD after at least two prior systemic therapies were randomized 1:1:1 to receive intravenous axatilimab at 0.3 mg/kg every 2 weeks (n=80), 1 mg/kg every 2 weeks (n=81), or 3 mg/kg every 4 weeks (n=80). Randomization was stratified by chronic GVHD severity and prior use of FDA-approved therapies (ibrutinib, ruxolitinib, or belumosudil). The primary endpoint was overall response rate (complete or partial response) within the first six cycles. The key secondary endpoint was a patient-reported reduction in symptom burden, defined as a decrease of more than 5 points on the modified Lee Symptom Scale (range 0–100).

Results: The overall response rate was 74% (95% CI, 63%–83%) in the 0.3 mg/kg group, 67% (95% CI, 55%–77%) in the 1 mg/kg group, and 50% (95% CI, 39%–61%) in the 3 mg/kg group, exceeding the predefined efficacy threshold in all groups. A clinically meaningful reduction in symptom burden was reported in 60%, 69%, and 41% of patients in the respective dose groups. Median time to response was less than 2 months across all groups. Organ-specific responses were observed in all affected organs, including skin, lungs, joints, and fascia.

The most common adverse events were dose-dependent transient laboratory abnormalities related to CSF1R blockade, such as elevations in liver enzymes and creatine kinase, which were not associated with clinical symptoms or end-organ damage. Periorbital edema occurred more frequently at higher doses. Adverse events leading to discontinuation occurred in 6% of patients in the 0.3 mg/kg group, 22% in the 1 mg/kg group, and 18% in the 3 mg/kg group. Serious infections were reported but were not dose-dependent.

Conclusions: Axatilimab demonstrated significant efficacy in patients with heavily pretreated recurrent or refractory chronic GVHD, with the highest response rates and best tolerability observed at the lowest dose tested (0.3 mg/kg every 2 weeks). Targeting CSF1R-dependent monocytes and macrophages may represent a novel therapeutic strategy in chronic GVHD.

Implications for Practice: Axatilimab offers a potential new treatment option for patients with chronic GVHD refractory to standard therapies, including those who have failed prior FDA-approved treatments. Clinicians should consider axatilimab as a therapeutic option while monitoring for transient laboratory abnormalities associated with CSF1R blockade. The lower dose appears to provide optimal efficacy with fewer adverse events.

Study Strengths and Limitations: Strengths include the randomized, multinational design and inclusion of patients with severe, refractory chronic GVHD who had received multiple prior therapies. Limitations include the lack of a comparator group, which may introduce outcome-reporting bias, and the small sizes of subgroups, limiting the generalizability of certain findings.

Future Research: Further studies are needed to confirm these results, assess long-term outcomes, and explore axatilimab in earlier lines of therapy and in combination with other treatments. Investigations into the use of axatilimab in other autoimmune diseases characterized by CSF1R-driven macrophage-mediated inflammation and fibrosis are also warranted.

Reference: Wolff D, Cutler C, Lee SJ, Pusic I, Bittencourt H, White J, Hamadani M, et al. Axatilimab in Recurrent or Refractory Chronic Graft-versus-Host Disease. N Engl J Med. 2024. DOI: http://doi.org/10.1056/NEJMoa2401537

 


RCT: Pembrolizumab Plus Chemotherapy Improved Overall Survival in Early-Stage Triple-Negative Breast Cancer

18 Sep, 2024 | 16:08h | UTC

Background: Early-stage triple-negative breast cancer (TNBC) is an aggressive subtype with limited treatment options and poor prognosis. The phase 3 KEYNOTE-522 trial previously demonstrated that adding pembrolizumab to chemotherapy improved pathological complete response rates and event-free survival in this population.

Objective: To determine whether neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab improves overall survival compared to neoadjuvant chemotherapy alone in patients with early-stage TNBC.

Methods: In this multicenter, randomized, double-blind, placebo-controlled phase 3 trial (KEYNOTE-522), 1174 patients with previously untreated stage II or III TNBC were randomized 2:1 to receive neoadjuvant pembrolizumab (200 mg every 3 weeks) plus chemotherapy (paclitaxel and carboplatin, followed by doxorubicin–cyclophosphamide or epirubicin–cyclophosphamide) or placebo plus the same chemotherapy regimen. After surgery, patients received adjuvant pembrolizumab or placebo every 3 weeks for up to nine cycles. The primary endpoints were pathological complete response and event-free survival; overall survival was a key secondary endpoint.

Results: After a median follow-up of 75.1 months, the estimated 5-year overall survival was 86.6% in the pembrolizumab–chemotherapy group versus 81.7% in the placebo–chemotherapy group (hazard ratio for death, 0.66; P=0.002). The 5-year event-free survival was 81.2% versus 72.2%, respectively (hazard ratio for event or death, 0.65; 95% CI, 0.51–0.83). Grade 3 or higher treatment-related adverse events occurred in 77.1% of patients receiving pembrolizumab–chemotherapy and 73.3% receiving placebo–chemotherapy. Serious treatment-related adverse events occurred in 34.1% and 20.1% of patients, respectively.

Conclusions: Neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab significantly improved overall survival compared to chemotherapy alone in patients with early-stage TNBC.

Implications for Practice: The addition of pembrolizumab to standard neoadjuvant chemotherapy, followed by adjuvant pembrolizumab, should be considered a new standard of care for patients with high-risk, early-stage TNBC, offering a significant survival benefit.

Study Strengths and Limitations: Strengths include the large, international, randomized design, the use of a placebo control, and long-term follow-up. Limitations include the inability to isolate the effects of neoadjuvant versus adjuvant pembrolizumab and the exclusion of adjuvant capecitabine from the treatment protocol.

Future Research: Further studies should focus on identifying biomarkers predictive of response to pembrolizumab, optimizing the sequencing and duration of immunotherapy, and evaluating the addition of other agents to improve outcomes in early-stage TNBC.

Reference: Schmid P, et al. Overall Survival with Pembrolizumab in Early-Stage Triple-Negative Breast Cancer. N Engl J Med. 2024. DOI: http://doi.org/10.1056/NEJMoa2409932

 


RCT: Perioperative Durvalumab Plus Neoadjuvant Chemotherapy Improved Survival in Muscle-Invasive Bladder Cancer

18 Sep, 2024 | 15:22h | UTC

Background: Neoadjuvant cisplatin-based chemotherapy followed by radical cystectomy is the standard treatment for cisplatin-eligible patients with muscle-invasive bladder cancer (MIBC). Despite this approach, approximately 50% of patients experience recurrence within 3 years. Combining immunotherapy with chemotherapy may enhance outcomes, and durvalumab, a PD-L1 inhibitor, has shown promise when added to chemotherapy in previous studies.

Objective: To evaluate whether perioperative durvalumab combined with neoadjuvant gemcitabine–cisplatin improves event-free survival (EFS) and overall survival (OS) compared to neoadjuvant chemotherapy alone in cisplatin-eligible patients with operable MIBC.

Methods: In the phase 3, open-label, randomized NIAGARA trial, 1063 cisplatin-eligible patients with MIBC (clinical stage T2-T4aN0-1M0) were randomized 1:1 to receive either perioperative durvalumab plus neoadjuvant gemcitabine–cisplatin followed by radical cystectomy and adjuvant durvalumab (durvalumab group, n=533) or neoadjuvant gemcitabine–cisplatin followed by radical cystectomy alone (comparison group, n=530). The primary endpoints were EFS and pathological complete response (pCR). The key secondary endpoint was OS.

Results: At a median follow-up of 42.3 months, the estimated EFS at 24 months was 67.8% (95% CI, 63.6–71.7) in the durvalumab group versus 59.8% (95% CI, 55.4–64.0) in the comparison group (hazard ratio [HR] for progression, recurrence, not undergoing cystectomy, or death, 0.68; 95% CI, 0.56–0.82; P<0.001). The estimated OS at 24 months was 82.2% (95% CI, 78.7–85.2) in the durvalumab group versus 75.2% (95% CI, 71.3–78.8) in the comparison group (HR for death, 0.75; 95% CI, 0.59–0.93; P=0.01). Grade 3 or 4 treatment-related adverse events occurred in 40.6% of patients in the durvalumab group and 40.9% in the comparison group. Radical cystectomy was performed in 88.0% of patients in the durvalumab group and 83.2% in the comparison group.

Conclusions: Perioperative durvalumab combined with neoadjuvant chemotherapy significantly improved event-free and overall survival compared to neoadjuvant chemotherapy alone in cisplatin-eligible patients with operable MIBC.

Implications for Practice: Adding perioperative durvalumab to neoadjuvant chemotherapy may represent a new option for cisplatin-eligible patients with MIBC, offering improved survival outcomes without increasing significant adverse events or delaying surgery.

Study Strengths and Limitations: Strengths include the large sample size and randomized, phase 3 design. Limitations encompass the open-label design and inability to isolate the effects of neoadjuvant versus adjuvant durvalumab. Additionally, the trial was conducted before the widespread use of adjuvant nivolumab, potentially affecting the generalizability of the results.

Future Research: Further studies are needed to delineate the relative contributions of neoadjuvant and adjuvant durvalumab and to explore biomarkers such as circulating tumor DNA to guide treatment decisions.

Reference: Powles T, Catto JWF, Galsky MD, et al. Perioperative Durvalumab with Neoadjuvant Chemotherapy in Operable Bladder Cancer. N Engl J Med. 2024 Sep 15. DOI: http://doi.org/10.1056/NEJMoa2408154

 


RCT: Nivolumab Plus Ipilimumab Shows Sustained 10-Year Survival Benefit in Advanced Melanoma

18 Sep, 2024 | 15:06h | UTC

Background: Advanced melanoma historically had a poor prognosis, with median survival under 12 months before 2011. The advent of immune checkpoint inhibitors like nivolumab (anti–PD-1) and ipilimumab (anti–CTLA-4) has significantly improved outcomes. Previous results from the CheckMate 067 trial showed longer overall survival with nivolumab plus ipilimumab or nivolumab alone compared to ipilimumab alone. As patients now live beyond 7.5 years, longer-term data are needed to address new clinical questions about survival and disease progression.

Objective: To assess the final 10-year outcomes of overall survival, melanoma-specific survival, and response durability in patients with advanced melanoma treated with nivolumab plus ipilimumab, nivolumab monotherapy, or ipilimumab monotherapy.

Methods: In the phase 3 CheckMate 067 trial, 945 patients with untreated, unresectable stage III or IV melanoma were randomized 1:1:1 to receive:

  • Nivolumab plus ipilimumab: Nivolumab (1 mg/kg) and ipilimumab (3 mg/kg) every 3 weeks for four doses, then nivolumab (3 mg/kg) every 2 weeks.
  • Nivolumab monotherapy: Nivolumab (3 mg/kg) every 2 weeks plus placebo.
  • Ipilimumab monotherapy: Ipilimumab (3 mg/kg) every 3 weeks for four doses plus placebo.

Treatment continued until disease progression, unacceptable toxicity, or withdrawal of consent. Randomization was stratified by BRAF mutation status, metastasis stage, and PD-L1 expression. Primary endpoints were overall survival and progression-free survival; secondary endpoints included objective response rates and subgroup analyses.

Results: After 10 years, median overall survival was:

  • 71.9 months with nivolumab plus ipilimumab,
  • 36.9 months with nivolumab,
  • 19.9 months with ipilimumab.

Hazard ratios for death were 0.53 (95% CI, 0.44–0.65; P<0.001) for nivolumab plus ipilimumab vs. ipilimumab, and 0.63 (95% CI, 0.52–0.76; P<0.001) for nivolumab vs. ipilimumab. Ten-year overall survival rates were 43% with combination therapy, 37% with nivolumab, and 19% with ipilimumab. Median melanoma-specific survival was not reached (>120 months) with combination therapy (37% alive at study end), 49.4 months with nivolumab, and 21.9 months with ipilimumab. Among patients alive and progression-free at 3 years, 10-year melanoma-specific survival was 96% with combination therapy, 97% with nivolumab, and 88% with ipilimumab. No new safety signals were observed over the extended follow-up.

Conclusions: Nivolumab plus ipilimumab demonstrated a sustained 10-year survival benefit over ipilimumab monotherapy in advanced melanoma. Nivolumab monotherapy also improved survival compared to ipilimumab, though the combination provided the greatest benefit.

Implications for Practice: These 10-year results support nivolumab plus ipilimumab as a preferred first-line treatment for advanced melanoma, offering potential for long-term survival and possible cure. Clinicians should balance improved efficacy against higher adverse event rates with combination therapy and monitor patients accordingly.

Study Strengths and Limitations: Strengths include the large, randomized, multicenter design and extended follow-up, providing robust survival data. Limitations include the trial not being powered for formal comparison between combination and monotherapy, and potential confounding from subsequent therapies on long-term outcomes.

Future Research: Further studies should aim to identify biomarkers predicting long-term response, optimize patient selection for combination therapy, and develop treatments for patients unresponsive to current immune checkpoint inhibitors.

Reference: Wolchok JD, et al. (2024). Final, 10-Year Outcomes with Nivolumab plus Ipilimumab in Advanced Melanoma. New England Journal of Medicine. 2024. DOI: http://doi.org/10.1056/NEJMoa2407417

 


Stay Updated in Your Specialty

Telegram Channels
Free

WhatsApp alerts 10-day free trial

No spam, just news.