Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
ObjectiveIn this study, we aimed to perform a network meta-analysis to compare the effectiveness of NAs in decreasing the reactivation of HBV, reducing chemotherapy disruption, and improving survival in oncology patients.MethodsRelevant randomized controlled trials (RCT) evaluating the impact of NAs in HBV infected-related oncology patients were retrieved from electronic databases. The outcome indicators included reactivation rate, survival rate of 1 to 3 years after treatment, and chemotherapy disruption rate. The studies were evaluated for bias using the RCT risk of bias assessment tool recommended in the Cochrane Handbook. The risk ratio (RR) was used to compare the outcome indicators for the anti-viral treatment, and the surface under the cumulative ranking curves (SUCRA) was used to identify the optimal therapeutic regime.ResultsA total of 67 trials containing 5722 patients were included in this study. Regarding the reduction of reactivation rate, entecavir, lamivudine, adefovir alone were less effective than the combination of lamivudine and entecavir (94.9%), with RR values ranging from 3.16 to 3.73. However, based on SUCRA, the efficacy of telbivudine (80.3%) and the combination of lamivudine and adefovir dipivoxil (58.8%) were also acceptable. Entecavir (RR values ranging from 1.25 to 1.50) and lamivudine (RR values ranging from 1.27 to 1.35) can prolong the survival rate of patients at 1-3 years, and were better than adefovir dipivoxil in the comparison of 1-year survival rate. The RR values were 1.18 and 1.19, respectively. And entecavir ‘s ranking in SUCRA was more stable. Entecavir, lamivudine, and tenofovir all reduced chemotherapy interruption rates compared with no antiviral therapy, especially for tenofovir.ConclusionsCurrent evidence shows that lamivudine combined with entecavir, telbivudine, and lamivudine combined with adefovir dipivoxil were the most effective in preventing virus reactivation in HBV infected-related cancer patients treated with chemotherapy. Entecavir had the most stable effect on survival, while tenofovir had the best impact on reducing the chemotherapy disruption rate. Due to limited quality and quantity of the included studies, more high-quality studies are required to verify the above conclusions.Systematic review registrationPROSPEROI [https://www.crd.york.ac.uk/PROSPERO/index.php], identifier CRD4202250685.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
BackgroundAdvanced triple-negative breast cancer (TNBC) presents significant therapeutic challenges, particularly in Asian populations, which exhibit distinct biological and genetic characteristics. Immunotherapy combined with chemotherapy has emerged as a promising approach; however, its efficacy compared to chemotherapy alone remains under investigation. This meta-analysis aims to evaluate the clinical outcomes of PD-1/PD-L1 inhibitors combined with chemotherapy (PIC) versus chemotherapy alone in the treatment of advanced TNBC in Asian patients.MethodsA systematic literature search was performed across six databases for phase 3 randomized controlled trials (RCTs). Only studies comparing the outcomes of PIC versus chemotherapy alone in patients with advanced TNBC, including subgroup analyses of Asian populations, were included. Data were pooled to assess overall survival (OS), progression-free survival (PFS), responses, and safety profiles.ResultsA total of 1041 patients from five phase 3 RCTs were included in the final analysis. Compared to chemotherapy alone, PIC therapy significantly improved PFS (hazard ratio [HR]: 0.74 [0.62, 0.88], P = 0.0008). No significant difference was observed in OS (HR: 0.78 [0.55, 1.12], P = 0.18), although a slight trend favoring PIC therapy was noted. Among PD-L1-positive patients, both OS (HR: 0.62 [0.44, 0.86], P = 0.005) and PFS (HR: 0.66 [0.50, 0.86], P = 0.003) were significantly improved in the PIC group. The PIC group also exhibited a substantially higher OS rate at 12–36 months and a higher PFS rate at 6–30 months. However, the incidence of immune-related AEs (irAEs) (risk ratio [RR]: 1.69 [1.33, 2.15], P < 0.0001) and grade 3–5 irAEs (RR: 3.11 [1.59, 6.10], P = 0.001) was significantly higher in the PIC group. The most common irAEs in the PIC group were hypothyroidism (14.40%), dermatitis (10.00%), and infusion reactions (8.85%). Both treatment groups exhibited similar response rates and treatment-related AEs (TRAEs).ConclusionsIn Asian patients with advanced TNBC, PIC significantly improved survival compared to chemotherapy alone. Although the combination therapy was associated with a higher incidence of irAEs, its clinical benefits support its use as a viable treatment option for this population.Systematic review registrationhttps://www.crd.york.ac.uk/prospero/, identifier CRD42024622428.
https://www.cognitivemarketresearch.com/privacy-policyhttps://www.cognitivemarketresearch.com/privacy-policy
According to Cognitive Market Research, the global Metastatic Bones Cancer Medication Treatment Market size will be USD 1514.2 million in 2024. It will expand at a compound annual growth rate (CAGR) of 5.20% from 2024 to 2031.
North America held the major market share for more than 40% of the global revenue with a market size of USD 605.68 million in 2024 and will grow at a compound annual growth rate (CAGR) of 7.2% from 2024 to 2031.
Europe accounted for a market share of over 30% of the global revenue with a market size of USD 454.26 million.
Asia Pacific held a market share of around 23% of the global revenue with a market size of USD 348.27 million in 2024 and will grow at a compound annual growth rate (CAGR) of 11.0% from 2024 to 2031.
Latin America had a market share of more than 5% of the global revenue with a market size of USD 75.71 million in 2024 and will grow at a compound annual growth rate (CAGR) of 8.4% from 2024 to 2031.
Middle East and Africa had a market share of around 2% of the global revenue and was estimated at a market size of USD 30.28 million in 2024 and will grow at a compound annual growth rate (CAGR) of 8.7% from 2024 to 2031.
The chemotherapy category is the fastest growing segment of the Metastatic Bones Cancer Medication Treatment industry
Market Dynamics of Metastatic Bones Cancer Medication Treatment Market
Key Drivers for Metastatic Bones Cancer Medication Treatment Market
Growing Cancer Awareness Campaigns Fueling Demand for Metastatic Bone Disease
An increase in cancer awareness initiatives is helping to enhance patient knowledge, which in turn is resulting in earlier screenings and diagnoses. The patient's chances of survival may significantly improve with early cancer detection and diagnosis. According to recent medical study, early detection (at stage one) of most malignancies increases their survival percentage at one and five years. Early detection and diagnosis of the disease are aided by governments' and other stakeholders' persistent efforts to promote self-screening and raise awareness of the many forms of cancer. Campaigns like "Race for Life" and "Stand Up to Cancer" are run by Cancer Research UK, and "Great American Smokeout" and "Relay for Life" are run by the American Cancer Society. Campaigns like "Race for Life" and "Stand Up to Cancer" are run by Cancer Research UK, and "Great American Smokeout" and "Relay for Life" are run by the American Cancer Society. These efforts have improved survival rates, collected money for research, and encouraged good diet and lifestyle choices.
Rise in Prevalence of Cancer to Drive Market Growth
The rise in the prevalence of cancer is a significant driver of the metastatic bone cancer medication treatment market. As cancer incidence increases globally, more patients develop bone metastases, particularly from common cancers such as breast, prostate, and lung cancer. This growing patient population fuels demand for advanced treatments targeting bone metastasis. Additionally, improved diagnostic techniques, such as advanced imaging and biomarker testing, allow for earlier detection and better management of metastatic bone cancer, further contributing to market growth. The increasing awareness of cancer treatments and the availability of new, more effective therapies, such as bone-modifying agents, targeted therapies, and immunotherapies, are helping improve patient outcomes and driving market expansions.
Restraint Factor for the Metastatic Bones Cancer Medication Treatment Market
Expensive therapies and medications, will Limit Market Growth
Expensive therapies and medications are a major restraint in the metastatic bone cancer medication treatment market. Advanced treatments like targeted therapies, immunotherapies, and bone-modifying agents often come with high costs, which can be prohibitive for patients, especially in low-income or developing regions. Additionally, the cost of these therapies places a significant burden on healthcare systems, potentially limiting access to effective treatment. Insurance coverage can also vary, further restricting patient access to necessary medications. These financial barriers can delay diagnosis and treatment, leading to poorer outcomes. As a result, affordability remains a critical challenge, hindering the widespread adoption of innovative treatments...
Glioblastoma Multiforme Treatment Market Size 2024-2028
The glioblastoma multiforme treatment market size is forecast to increase by USD 1.36 billion at a CAGR of 8.2% between 2023 and 2028.
The glioblastoma multiforme (GBM) treatment market is experiencing significant growth due to the rising incidence of this aggressive brain cancer and increasing research and development activities In the healthcare sector. According to the American Brain Tumor Association's Cancer Observatory, GBM accounts for approximately 15% of all primary brain tumors and is the most common malignant brain tumor in adults. Current treatment options include surgery, targeted therapy, and chemotherapy. Surgery remains the primary treatment modality for GBM, with various options such as awake craniotomy and awake craniotomy with intraoperative monitoring. This includes various strategies such as checkpoint inhibitors, monoclonal antibodies, and gene therapy. However, these procedures come with adverse effects such as cognitive impairment and motor deficits. Targeted therapies, such as temozolomide, are used in conjunction with surgery and chemotherapy to improve patient outcomes. The pharmaceutical supply chain plays a crucial role in ensuring the timely availability of these treatments to patients. Despite advancements in treatment options, the high mortality rate associated with GBM necessitates continued research and development efforts to improve patient outcomes and quality of life.
What will be the Size of the Glioblastoma Multiforme Treatment Market During the Forecast Period?
Request Free Sample
Glioblastoma multiforme (GBM), a malignant tumor of the central nervous system (CNS), is a significant health concern worldwide. According to the Global Cancer Observatory, CNS cancers accounted for approximately 2.3% of all cancer diagnoses and 2.5% of cancer deaths in 2020. This article provides an overview of the current treatment landscape for GBM, focusing on surgical, radiation, chemotherapy, targeted therapy, immunotherapy, and emerging novel therapies. Surgery is the primary treatment modality for GBM, with the primary goal being to remove as much of the tumor as possible while minimizing damage to healthy brain tissue. Ambulatory surgical centers and hospitals offer various surgical procedures, including craniotomy and awake craniotomy. Following surgery, patients typically undergo radiation therapy to target any remaining cancer cells. External beam radiation therapy (EBRT) is the most common approach, delivering high-energy radiation to the tumor site. Chemotherapy is often used in combination with radiation therapy to enhance the therapeutic effect.
Moreover, temozolomide, a chemotherapeutic agent, is the most widely used drug for GBM treatment. Corticosteroids, such as dexamethasone, are also frequently administered to reduce swelling and improve symptoms. Targeted therapies, such as bevacizumab, are designed to specifically target the molecular mechanisms of GBM. These therapies inhibit the growth and spread of cancer cells by blocking the action of specific proteins. Moreover, combination Therapies: Combination therapies, which involve the use of multiple treatment modalities, are increasingly being explored to improve treatment outcomes for GBM. For example, the combination of temozolomide and radiation therapy has been shown to improve survival rates compared to monotherapy. Immunotherapy: Immunotherapy, which harnesses the power of the immune system to fight cancer, is an emerging treatment approach for GBM. Personalized Medicine: The heterogeneity of GBM necessitates personalized treatment approaches.
Thus, drug classes and treatment procedures are being tailored to individual patients based on their unique tumor characteristics, such as genetic mutations and protein expression profiles. The treatment market for GBM is continually evolving, with a focus on developing novel therapies and combination strategies to improve patient outcomes. The pharmaceutical supply chain plays a crucial role in ensuring the availability and accessibility of these treatments. As the understanding of GBM biology deepens, so too will the range and effectiveness of treatment options.
How is this Glioblastoma Multiforme Treatment Industry segmented and which is the largest segment?
The glioblastoma multiforme treatment industry research report provides comprehensive data (region-wise segment analysis), with forecasts and estimates in 'USD billion' for the period 2024-2028, as well as historical data from 2018-2022 for the following segments.
End-user
Hospitals
Clinics
Ambulatory surgical centers
Geography
North America
Canada
US
Europe
Germany
UK
Asia
China
Rest of World (ROW)
By End-user Insights
The hospitals segment is estimated to witness significant growth during the forecast period.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
BackgroundCost-effectiveness of atezolizumab, as a treatment for advanced non-small-cell lung cancer (NSCLC) patients who cannot receive a platinum-containing regimen,was still unknown. Our objective was to evaluate the cost-effectiveness of atezolizumab vs. chemotherapy in this indication from the perspective of UK healthcare system.MethodsFrom the global, randomised, open-label, phase III IPSOS trial, clinical inputs and patient characteristics were obtained. A partitioned survival model with three health states was built: Progression-free survival, progressed disease and death. A lifetime time horizon was applied, with an annual discount rate of 3.5%. Additionally, the willingness-to-pay threshold of £50,000/QALY was utilized. Primary outcomes were quality-adjusted life-year (QALY), costs, and incremental cost-effectiveness ratio (ICER). Sensitivity, scenario, and subgroup analyses were used to assess the reliability of base-case results. Price simulations were carried out in order to provide information for the pricing strategy at specific willingness-to-pay threshold.ResultsIn the base-case analysis, atezolizumab resulted in a gain of 0.28 QALYs and an ICER of £94,873/QALY compared to chemotherapy, demonstrating no cost-effectiveness. Price simulation results revealed that atezolizumab would be preferred at a price lower than £2,215 (a reduction of 41.8%) at the willingness-to-pay threshold of £50,000. Sensitivity, scenario and subgroup analyses revealed these conclusions were generally robust, the model was most sensitive to the price of atezolizumab and subsequent medication. Furthermore, atezolizumab was found to be more cost-effective for patients displaying a positive PD-L1 expression, with an ICER of £72,098/QALY as compared to chemotherapy.ConclusionAtezolizumab is not cost-effective for patients with advanced NSCLC ineligible for platinum-containing regimen, potential price reduction is necessary.
Breast Cancer Therapeutics Market Size 2024-2028
The breast cancer therapeutics market size is forecast to increase by USD 15.64 bn at a CAGR of 8.4% between 2023 and 2028.
The market is experiencing significant growth due to the rising prevalence of breast cancer and the emergence of novel therapeutics. Hormone therapy and radiation therapy continue to be popular treatment options, while oncology drugs such as monoclonal antibodies, biosimilars, and vaccines are gaining popularity. Artificial intelligence is revolutionizing cancer diagnostics and treatment plans, enabling early detection and personalized treatment. Online pharmacies and telemedicine are making treatments more accessible, especially in remote areas. The high cost of treatment remains a challenge, but education and nutrition programs are helping to mitigate this issue. Protein-based therapies, gelatin, and tablets are also key components of breast cancer therapeutics.In the US, the market is expected to grow steadily due to increasing awareness and advancements in technology and treatment methods.
What will be the Size of the Breast Cancer Therapeutics Market During the Forecast Period?
Request Free SampleThe market encompasses a range of treatments designed to combat this prevalent form of cancer among females. Key therapeutic approaches include chemotherapy, radiation therapy, hormone therapy, targeted therapy, and immunotherapy. Oncology drugs and therapeutics play a pivotal role in addressing breast cancer, targeting various proteins and cells, such as milk ducts, milk-producing lobules, and tumors. Breast cancer awareness initiatives and screening programs are instrumental in early detection, leading to improved patient outcomes. Artificial intelligence and advanced diagnostics are increasingly utilized to enhance the accuracy of diagnoses, enabling more effective interventions. Breast cancer can manifest as lumps, red patches, pain, swelling, or symptoms affecting the nipple, bones, breathing, or lymph nodes.Surgical tumor removal and medications, including hormonal therapies, are essential components of treatment plans. Despite advancements, breast cancer continues to pose significant health challenges, with ongoing research and innovation required to address the complexities of this disease.
How is this Breast Cancer Therapeutics Industry segmented and which is the largest segment?
The breast cancer therapeutics industry research report provides comprehensive data (region-wise segment analysis), with forecasts and estimates in 'USD billion' for the period 2024-2028, as well as historical data from 2018-2022 for the following segments. Disease TypeInvasive breast cancersDuctal carcinoma in situTherapyTargeted therapyHormonal therapyChemotherapyGeographyNorth AmericaCanadaUSEuropeGermanyUKAsiaChinaRest of World (ROW)
By Disease Type Insights
The invasive breast cancers segment is estimated to witness significant growth during the forecast period. Invasive ductal carcinoma, also known as infiltrating ductal carcinoma, is the most prevalent type of breast cancer, accounting for approximately 80% of diagnoses. This form of cancer arises when cancerous cells invade the fibrous or fatty tissue of the breast beyond the milk ducts. Symptoms include breast swelling, skin irritation, pain, nipple discomfort, redness, and nipple discharge, among others. Risk factors include genetics, hormonal imbalances, obesity, alcohol intake, and prior radiation therapy. Hormonal therapies, such as tamoxifen and aromatase inhibitors, chemotherapy, radiation therapy, targeted therapies, including Herceptin for HER2-positive tumors, and immunotherapies are common treatments. Early detection through breast cancer awareness campaigns, screening programs, and regular self-examinations contribute to improved survival probabilities and reduced morbidity.The development of oncology drugs and therapeutics, including proteins and biosimilars, continues to advance treatment options. Government support, hospital visits, and recovery rates are crucial aspects of breast cancer care. Hormonal therapy, obesity, and family history are significant factors in cancer type analysis. Hospital pharmacies and online pharmacies provide access to medications.
Get a glance at the market report of various segments Request Free Sample
The Invasive breast cancers segment was valued at USD 22.55 bn in 2018 and showed a gradual increase during the forecast period.
Regional Analysis
North America is estimated to contribute 46% to the growth of the global market during the forecast period. Technavio’s analysts have elaborately explained the regional trends and drivers that shape the market during the forecast period.
For more insights on the market size of various regions, Request Free Sample
The North American the market is experiencing significant growth due to the availability of r
Traditional surgery plus radiotherapy or chemotherapy, existing targeted therapies failed to significantly improve the survival rate of recurrent endometrial cancer, so suggesting that mechanism of recurrence and progression that modulates in endometrial cancer is clinically important. Here, we show that GPER(G protein-coupled estrogen receptor 1) was binded to AMF, and the complex were translocation form plasma to cytoplasmic. Mechanistic investigations elucidated that interaction of AMF with GPER triggers phosphoinositide-3-kinase (PI3K) signaling activating and accelerating the ability of endometrial cancer cells growth. Furthermore, we found that AMF may contribute to GPER-mediated endometrial cancer progression using animal experiments and human histological experiments which be consistent with the above conclusions. On the basis of these evidences including invivo and invitro, our findings suggest that AMF–GPER interaction might be novel key molecular targets for therapeutic management of patients with endometrial cancer, whose disease were progression and recurrence.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
ObjectiveTo compare cervical cancer recurrence and patient survival after radical hysterectomy followed by either adjuvant chemotherapy (AC) or adjuvant radiotherapy with or without concurrent chemotherapy (AR/CCRT).MethodsWe systematically searched PubMed, EMBASE, the Cochrane Library and clinicaltrials.gov to identify studies reporting recurrence or survival of cervical cancer patients who received AC or AR/CCRT after radical hysterectomy. Data were meta-analyzed using a random-effects model, and heterogeneity was evaluated using the I2 test. Subgroup and sensitivity analyses were performed to identify potential sources of heterogeneity.ResultsThe meta-analysis included 14 non-randomized studies and two randomized controlled trials, altogether involving 5,052 cervical cancer patients. AC and AR/CCRT groups did not differ significantly in rates of total or local recurrence or mortality. Nevertheless, AC was associated with significantly lower risk of distant recurrence [odds ratio (OR) 0.67, 95% confidence interval (CI) 0.55-0.81] and higher rates of overall survival [hazard ratio (HR) 0.69, 95%CI 0.54-0.85] and disease-free survival rate (HR 0.77, 95%CI 0.62-0.92).ConclusionsAC may be an effective alternative to AR/CCRT for cervical cancer patients after radical hysterectomy, especially younger women who wish to preserve their ovaries and protect them from radiation damage.Systematic Review Registrationhttps://www.crd.york.ac.uk/prospero/, identifier PROSPERO (CRD42021252518).
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
IntroductionWe did a systematic review and meta-analysis to assess the efficacy and safety of immune checkpoint inhibitors combined with or without chemotherapies in patients with esophageal squamous cell carcinoma.MethodsData related to the treatment of esophageal squamous cell carcinoma with immune checkpoint inhibitors therapy were retrieved from the database construction to August 2022. The risk of bias was assessed using the Cochrane Manual standard and RevMan 5.3 software for data synthesis. The outcome measures observed included overall survival, 12-month survival, disease control rate, objective response rate, treatment-related adverse events of grade 3 or higher, and progression-free survival. The adverse reactions included fatigue, diarrhea, hypothyroidism, rash, anemia, and anorexia.ResultsIn this meta-analysis, a total of 17 randomized controlled trials were included. In first-line therapy, immune checkpoint inhibitors combined with or without chemotherapy in the treatment of esophageal squamous cell carcinoma was more effective than chemotherapy alone. Overall survival, 12-month survival rate, and objective response rate were statistically significant. Among second-line treatments, immune checkpoint inhibitors combined with or without chemotherapy in the treatment of esophageal squamous cell carcinoma had statistically significant overall survival, 12-month survival, objective response rate, treatment-related adverse events of grade 3 or higher, and progression-free survival compared with chemotherapy alone.ConclusionBoth first- and second-line immune checkpoint inhibitors are effective for esophageal squamous cell carcinoma, and the adverse reactions are controllable and safe.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier CRD42021282586.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Background: Pelareorep is an oncolytic virus that causes oncolytic effects in many solid tumors, and it has shown therapeutic benefits. However, few studies have compared pelareorep combined with chemotherapy to traditional chemotherapy alone in advanced solid tumors. Consequently, we intended to evaluate the effectiveness and safety of pelareorep plus chemotherapy in this paper.Methods: We searched four databases including PubMed, Embase, Cochrane Library and Web of Science comprehensively for studies comparing pelareorep combined with chemotherapy to chemotherapy alone in the treatment of advanced solid tumors. The outcomes measures were 1-year overall survival (OS), 2-year OS, 4-month progression-free survival (PFS), 1-year PFS, objective response rate (ORR), any-grade adverse events (any-grade AEs), and severe AEs (grade ≥ 3).Results: There were five studies involving 492 patients included in the study. Combination therapy did not significantly improve clinical outcomes in terms of 1-year OS [RR = 1.02, 95%CI = (0.82–1.25)], 2-year OS [RR = 1.00, 95%CI = (0.67–1.49)], 4-month PFS [RR = 1.00, 95%CI = (0.67–1.49)], 1-year PFS [RR = 0.79, 95%CI = (0.44–1.42)], and ORR [OR = 0.79, 95%CI = (0.49–1.27)] compared to chemotherapy alone, and the subgroup analysis of 2-year OS, 1-year PFS, and ORR based on countries and tumor sites showed similar results. In all grades, the incidence of AEs was greater with combination therapy, including fever [RR = 3.10, 95%CI = (1.48–6.52)], nausea [RR = 1.19, 95%CI = (1.02–1.38)], diarrhea [RR = 1.87, 95%CI = (1.39–2.52)], chills [RR = 4.14, 95%CI = (2.30–7.43)], headache [RR = 1.46, 95%CI = (1.02–2.09)], vomiting [RR = 1.38, 95%CI = (1.06–1.80)] and flu-like symptoms [RR = 4.18, 95%CI = (2.19–7.98)]. However, severe adverse events did not differ significantly between the two arms.Conclusion: Pelareorep addition to traditional chemotherapy did not lead to significant improvements in OS, PFS, or ORR in advanced solid tumor patients, but it did partially increase AEs in all grades, with no discernible differences in serious AEs. Therefore, the combination treatment is not recommended in patients with advanced solid tumors.Systematic Review Registration:https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=400841, identifier CRD42023400841
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
ObjectiveIt remains unclear what the best second-line treatment is for patients with small-cell lung cancer sensitive to previous platinum-based chemotherapy.MethodsWe systematically screened randomized controlled trials from several online databases. The primary outcome was objective response rate (ORR), and the secondary outcomes were disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and hematological complications graded 3 to 5. The efficacy of included treatments was ranked by surface under the cumulative ranking curve (SUCRA) value.ResultsWe included eleven trials involving 1560 patients in quantitative analysis. Triple chemotherapy containing platinum (TP, combination of cisplatin, etoposide, and irinotecan) was associated with favorable ORR (intravenous topotecan vs TP; odds ratio: 0.13, 95% CI:0.03-0.63; SUCRA, 0.94) and PFS (vs intravenous topotecan; hazard ratio, 0.5; 95% CI: 0.25-0.99; SUCRA, 0.90). Belotecan ranked highest for OS (SUCRA, 0.90), while intravenous topotecan plus Ziv-aflibercept ranked highest for DCR (SUCRA, 0.75). TP was more likely to cause anemia and thrombocytopenia while intravenous topotecan plus Ziv-aflibercept resulted in most neutrocytopenia.ConclusionTP is the first recommendation for the second-line treatment of sensitive relapsed SCLC. TP achieved priority in ORR and PFS with the most frequent adverse effects in anemia and thrombocytopenia. For patients who cannot tolerate the hematological adverse effects of triple chemotherapy, amrubicin is an optional option. Amrubicin had relatively good ORR and PFS, accompanied by fewer hematological complications. The rechallenge of the platinum doublet is inferior to amrubicin in ORR, DCR, and PFS. Oral topotecan has a similar effect compared with IV topotecan, but oral topotecan was associated with slightly higher safety and less stress in nursing. Belotecan contributed to the best PFS with slightly better safety but was not ideal in other outcomes.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022358256.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
IntroductionTo investigate the effects of hepatic arterial infusion chemotherapy (HAIC) with or without systemic chemotherapy compared to systemic chemotherapy alone in patients with locally advanced hepatocellular carcinoma (HCC).MethodsFollowing a registered protocol (PROSPERO 2023 CRD42023386780 Available from: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023386780), a comprehensive search was performed using reputable databases and registries up to December 26, 2022, with no language, publication date, or status restrictions. Only randomized controlled trials (RCTs) investigating the effects of HAIC with or without systemic chemotherapy versus systemic therapy alone were included. The primary outcomes were overall survival (OS), progression-free survival (PFS), and adverse events. The secondary outcomes included the objective response rate (ORR) and disease control rate (DCR). A random-effects model was used, and the certainty of the evidence was rated using GRADE.ResultsSeven RCTs involving 1,010 patients were included. All trials utilized sorafenib as the comparator. Five trials (690 patients) compared HAIC plus sorafenib to sorafenib alone, while two trials (320 patients) compared HAIC to sorafenib. The results indicate that HAIC, with or without sorafenib, may increase OS, PFS, and ORR compared with sorafenib alone. HAIC may enhance DCR, but the evidence is very uncertain. Adverse events were comparable between HAIC plus sorafenib and sorafenib alone. However, adverse events might be decreased in HAIC alone.DiscussionHAIC with or without systemic chemotherapy may improve survival outcomes and response rates of patients with HCC. Since the current body of evidence is moderate to very low, more robust randomized trials are needed to confirm the efficacy of HAIC.Systematic Review Registrationhttps://www.crd.york.ac.uk/prospero/display_record.php?RecordID=386780, identifier CRD42023386780.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
BackgroundWith the emergence of new anti-angiogenic treatments and the ongoing updates to clinical guidelines, the effectiveness and safety of these agents in treating platinum-sensitive/resistant ovarian cancer (OC) are yet to be fully determined. Therefore, we conducted a meta-analysis to evaluate the efficacy and safety of anti-angiogenic drugs combined with chemotherapy (CT) for platinum-sensitive OC (PSOC) or platinum-resistant OC (PROC).MethodsA comprehensive literature search was conducted across several databases, including PubMed, Web of Science, Embase, and the Cochrane Library, encompassing all pertinent randomized controlled trials (RCTs) up to 31 May 2024. The primary outcomes for the meta-analysis were progression-free survival (PFS) and overall survival (OS), while the objective response rate (ORR), adverse events (AEs) of any grade, and grade ≥3 AEs were considered secondary endpoints. Data synthesis involved the computation of hazard ratio (HR), relative risk (RR), along with their 95% confidence interval (CI) and prediction interval (PI). Trial sequential analysis was carried out using STATA 12.0, R software 4.3.1, and TSA v0.9.5.10 Beta software.ResultsThis meta-analysis encompassed 15 RCTs. The overall analysis revealed that compared to CT alone (or plus placebo), anti-angiogenic drugs combined with CT significantly improved PFS (HR [95% CI] = 0.573 [0.518–0.633], 95% PI: 0.383-0.876) and ORR (RR [95% CI] = 1.362 [1.260–1.472], 95% PI: 0.824–2.251), but also increased the incidence of grade ≥3 AEs (RR [95% CI] = 1.115 [1.070–1.162], 95% PI: 0.870–1.422) in PSOC patients. For PROC patients, this combination therapy notably improved PFS (HR [95% CI] = 0.542 [0.475–0.619], 95% PI: 0.322–0.930), OS (HR [95% CI] = 0.752 [0.646–0.875], 95% PI: 0.554-0.997), and ORR (RR [95% CI] = 2.141 [1.702–2.694], 95% PI: 0.839–5.307), whilst simultaneously elevating the risk of grade ≥3 AEs (RR [95% CI] = 1.487 [1.216–1.819], 95% PI: 0.755–2.828).ConclusionOur research verified the advantages of combining anti-angiogenic agents with CT in enhancing PFS and ORR for patients with PSOC, and also confirmed improvements in PFS, OS, and ORR for those with PROC. It is crucial for medical practitioners to remain alert to the potential occurrence of AEs when implementing this combined therapeutic approach in a clinical milieu.Systematic Review Registration:https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024552010.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
ObjectiveTo compare the efficacy and safety of venetoclax (VEN) in combination with chemotherapy (chemo) versus chemo alone in the treatment of acute myeloid leukemia (AML).MethodTo compare the efficacy and/or safety of VEN+chemo versus chemotherapy alone for AML, PubMed, Embase, Web of Science, and the Cochrane Library were used to searching up to June 2023. Comparisons included complete remission (CR), CR with incomplete hematologic recovery (CRi), morphologic leukemia-free state (MLFS), overall response rate (ORR), and adverse events (AEs).ResultA total of 9 articles were included, including 3124 patients. The baseline characteristics between two patient groups were similar. The combined analysis showed that compared with the group receiving chemo alone, the VEN+chemo group exhibited higher rates of CR, CRi, MLFS and ORR. Additionally, the VEN+chemo group had longer event-free survival (EFS) and overall survival (OS) durations. The incidence rates of AEs and serious AEs (SAEs) were similar between the two groups, but the early 30-day mortality rate was lower in the VEN+chemo group than in the chemo alone group.ConclusionThe VEN+chemo therapy demonstrates significant efficacy and safety profile in AML patients. However, more prospective studies are needed in the future to provide more accurate and robust evidence for treatment selection in patients.Systematic Review Registrationhttps://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023439288, identifier CRD42023439288.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
BackgroundCombining PD-1/PD-L1 inhibitors with chemotherapy (PIC) is a standard first-line treatment for advanced non-small cell lung cancer (NSCLC). The addition of bevacizumab to this regimen (PD-1/PD-L1 inhibitors+bevacizumab+chemotherapy [PIBC]) remains controversial regarding its potential to enhance antitumor efficacy in clinical practice. This meta-analysis aims to compare the antitumor effectiveness and safety profiles of PIBC with PIC.MethodsWe systematically searched six databases to identify eligible RCTs. The primary outcomes were overall survival (OS) and progression-free survival (PFS), while the secondary outcomes included treatment responses and adverse events (AEs).ResultsThree RCTs (IMpower150, jRCT2080224500, and ORIENT-31) comprising a total of 1529 patients were analyzed. The PIBC regimen significantly improved PFS (hazard ratio [HR]: 0.76 [0.66, 0.87], P < 0.0001), objective response rate (risk ratio [RR]: 1.36 [1.22, 1.51], P < 0.00001), and disease control rate (RR: 1.06 [1.00, 1.12], P = 0.04). The PFS rates were also higher in the PIBC group at 6 and 18 months. Both groups showed similar results in terms of OS, 3–36 month OS rates, and total AEs. However, the PIBC group exhibited a higher incidence of grade 3–5 AEs, serious AEs, grade 3–5 treatment-related AEs (TRAEs) and serious TRAEs. The most frequent grade 3–5 AEs in the PIBC group included anorexia (36.40%), decreased neutrophil count (16.25%), neutropenia (13.50%), reduced white blood cell count (12.12%), and febrile neutropenia (9.42%).ConclusionsPIBC appears to be better than PIC for advanced NSCLC offering improved PFS and response rates (ORR and DCR). However, its higher incidence of AEs requires cautious attention.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/view/CRD42024559146, identifier CRD42024559146.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
BackgroundSystemic treatments, namely, either monotherapy or combination therapy, are commonly administered to patients with advanced or metastatic colorectal cancer (CRC). This study aimed to provide the complete efficacy and safety profiles and ranking of systemic therapies for the treatment of unresectable advanced or metastatic CRC.MethodsWe searched PubMed, Embase, the Cochrane Library, and ClinicalTrials.gov from inception until June 30, 2021, and also the bibliographies of relevant studies. Randomized controlled trials comparing two or more treatments, namely, at least capecitabine, 5-fluorouracil, leucovorin, irinotecan, bevacizumab, cetuximab, oxaliplatin, or panitumumab were investigated. A network meta-analysis using the Bayesian approach was performed to compare the efficacy and safety of treatments. The surface under the cumulative ranking curve (SUCRA) was calculated for the probability of each treatment as the most effective. The overall response rate (ORR), disease control rate (DCR), overall survival (OS), progression-free survival (PFS), adverse events (AEs) grade ≥3, and serious adverse events (SAEs) were evaluated.ResultsOne hundred two publications with 36,147 participants were assigned to 39 different treatments. Among 11 treatments with full information on six outcomes, FOLFIRI/FOLFOX/FOLFOXIRI + bevacizumab significantly improved both the ORR and DCR, compared to FOLFIRI. Although FOLFOX and FOLFIRI/FOLFOX + cetuximab significantly prolonged both OS and PFS, treatments were comparable in terms of AEs grade ≥3 and SAEs. The top highest SUCRA values were observed in the FOLFOXIRI + panitumumab group for ORR (96%) and DCR (99%), FOLFIRI + bevacizumab + panitumumab group for OS (62%) and PFS (54%), and FOLFOXIRI + bevacizumab group for AEs grade ≥3 (59%) and SAEs (59%) outcomes.ConclusionsThese findings suggest an available range of systemic treatment therapies with different efficacy and safety profiles with patients. Further investigations of the side effects and mutation status are required to confirm our findings.Systematic Review Registrationhttps://www.crd.york.ac.uk/prospero/, identifier CRD42019127772
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
ObjectiveTo evaluate the efficacy and safety of PD-1/L1 inhibitors as first-line therapy in metastatic colorectal cancer(mCRC).MethodArticles evaluating first-line PD-1/L1 inhibitors for mCRC were sought in four databases (Pubmed, Embase, Web of Science, and the Cochrane Library) from the inception of the databases until 11 November 2023. Meta-analyses were conducted to assess the rates of progression-free survival (PFS), overall survival (OS), complete response (CR), partial response (PR), stable disease (SD), progressive disease (PD), objective response rate (ORR), disease control rate (DCR), and grade ≥ 3 treatment-related adverse events (trAEs).ResultsTotally nine studies were included for meta-analysis. A subgroup analysis was performed based on mismatch repair(MMR) status and regimens. In patients diagnosed with mismatch repair-deficient(dMMR) mCRC who received PD-1/L1 inhibitors as their first-line treatment, the ORR was 0.54 (95% CI, 0.39 to 0.68), the median PFS was 53.2 months, the Grade≥ 3 TRAEs rate was 0.33(95% CI, 0.12 to 0.60) and the median OS was not determined. For patients with proficient mismatch repair (pMMR) mCRC who underwent a combined treatment of PD-1/L1 inhibitors, anti-VEGF monoclonal antibody and chemotherapy as their first-line therapy, the ORR was 0.62 (95% CI, 0.56 to 0.68), the median PFS was 10.1 months, the median OS was 26.7 months, and the Grade≥ 3 TRAEs rate was 0.59(95% CI, 0.39 to 0.77).ConclusionOur results revealed that the utilization of PD-1/L1 inhibitors as first-line therapy for dMMR mCRC yielded highly favorable outcomes, while maintaining an acceptable level of safety. Administering a combination of PD-1/L1 inhibitors, anti-VEGF monoclonal antibody, and chemotherapy as first-line treatment in patients with pMMR mCRC led to an improved ORR. However, there was no significant improvement in the long-term prognosis of the tumor.Systematic review registrationhttps://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42024506196, identifier CRD42024506196.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Randomized controlled trials (RCTs) have assessed the efficacy of anti-programmed cell death 1 (PD- 1)/programmed cell death ligand 1 (PD-L1), alone or combined with other therapies, for ovarian cancer. However, the optimal strategy remains unclear. This study evaluated their effectiveness as monotherapy and in combination. We conducted a systematic review and Frequentist network meta-analysis (NMA) by searching PubMed, ScienceDirect, Web of Science, Scopus, Cochrane Library, and Clinicaltrials.gov databases. This analysis included RCTs comparing PD-L1/PD-1 inhibitors, alone or in combination with other therapies. Six studies involving 3,895 patients and eight treatment combinations were included. PD1/PDL1 inhibitors plus chemotherapy and PD1/PDL1 inhibitors plus ipilimumab showed the greatest progression-free survival (PFS) benefit (hazard ratio (HR) = 0.82, 95% confidence interval [CI]: 0.52–1.07; HR = 0.82, 95% CI:0.51–1.33) and overall survival (OS) (HR = 0.85, 95% CI:0.64–1.14; HR = 0.83, 95% CI:0.45–1.54). These combinations also improved overall response rates (ORR), especially with chemotherapy (OR = 3.06; 95% CI:1.42–6.60). Subgroup analysis suggested that PD-1/PD-L1 inhibitors plus chemotherapy provided the best PFS and OS in PD-L1-positive patients. Combining PD-1/PD-L1 inhibitors with chemotherapy or ipilimumab improved survival in ovarian cancer. However, PD-L1 expression may be a valuable biomarker for predicting the efficacy of PD-1/PD-L1 checkpoint inhibitors. This systematic review was preregistered in PROSPERO (CRD42022342057). Available from: https://www.crd.york.ac.uk/PROSPERO/view/CRD42022347967. Ovarian cancer is a serious disease that is often discovered too late. In recent years, researchers have developed new treatments called immune checkpoint inhibitors, including programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors. These medicines help the body’s immune system fight cancer more effectively. Although they work well in other types of cancer, it is still unclear how effective they are in ovarian cancer.This study looked at six clinical trials that tested these treatments in women with ovarian cancer. Researchers have compared different combinations to determine which one works best. They found that combining immune checkpoint inhibitors with chemotherapy or with another medicine called ipilimumab helped patients live longer and slowed the growth of the cancer. These combinations worked better than chemotherapy alone.Women whose tumors had high levels of a protein called PD-L1 showed even better results. These findings indicate that testing for PD-L1 could help doctors choose the best treatment for each patient. Overall, this study suggests that combining immune-based treatments with other therapies could help some women with ovarian cancer. The results are promising, but more research is needed.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
BackgroundMany options for third-line treatment of advanced gastric cancer (GC) or gastroesophageal junction carcinoma (GEJC) have been developed. Therapies including immunotherapy (nivolumab), chemotherapy (irinotecan, FTD/TPI), targeted therapy (apatinib), and antibody drug conjugates (ADC) have shown to increase the survival rates in patients, but few studies have compared the relative efficacy of these treatments. Here, we compared the efficacies of these regimens using network meta-analysis (NMA) to provide guides in selecting the best regimen and formulating a precise individualized treatment plan.MethodsThe published RCTs of phase II/III in PubMed, the Cochrane Central Register of Controlled Trials, and Embase were searched. The median overall survival (mOS) was the primary outcome of NMA, and the other outcomes were median progression-free survival (mPFS), disease control rate (DCR) (proportion of patients with confirmed CR, PR, or stable disease (SD)) and incidence of grade 3 or above adverse events (≥3AEs).ResultsFive phase II/III RCTs involving 1674 patients and 7 treatment regimens were analyzed. It showed that Trastuzumab Deruxtecan (DS-8201) prolonged the OS of patients significantly comparing with chemotherapy (HR: 0.59; 95% CI: 0.39-0.89) for the overall population. DS-8201 (HR: 0.27; 95% CI: 0.17-0.42) and chemotherapy (HR: 0.57; 95% CI: 0.47-0.7) improved the PFS significantly over nivolumab. Apatinib (RR: 3.04; 95% CI: 1.65-5.95) and DS-8201 (RR: 2.67; 95% CI: 1.51-4.83) were more effective than nivolumab in improving DCR. DS-8201 achieved greater OS benefits compared to chemotherapy (HR: 0.59; 95% CI: 0.39-0.88) for patients who were HER2-positive. We ranked the Bayesian surface under the cumulative ranking curve according to OS benefit, and showed that ADC ranked first for the general patient population and for patients with a HER2-positive diagnosis, intestinal histopathology, previous gastrectomy history, gastric origination cancer, ages over 65 and ECOG PS=0/1, followed by nivolumab and apatinib. For patients with GEJC, nivolumab ranked first.ConclusionsNivolumab, apatinib, chemotherapy, and ADC all improved the OS of GC/GEJC patients significantly. ADC may be the best option for the overall population of GC, as well as for patients with HER2-overexpression, intestinal histopathology, previous gastrectomy history, gastric origination cancer, ages over 65 and ECOG PS=0/1, followed by nivolumab and apatinib. Nivolumab may be the first treatment option for GEJC patients.Systematic review registrationhttps://www.crd.york.ac.uk/prospero, identifier CRD42022364714.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
BackgroundCombining epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) with chemotherapy (ETC) offers more advantages for patients with EGFR-positive non-small cell lung cancer (NSCLC) than using EGFR TKIs alone (ET). However, whether this conclusion applies to patients with brain metastases (BM) remains controversial. This meta-analysis was performed to evaluate the benefits and risks of the two groups.MethodsSix databases were systematically searched for relevant literatures comparing ETC versus ET in treating EGFR-positive NSCLC patients with BM. The primary outcome assessed was overall survival (OS), while secondary outcomes included progression-free survival (PFS), and central nervous system (CNS)-PFS, responses, progression status and safety.ResultsSeven studies based on five randomized clinical trials with 550 patients were included. The ETC group exhibited better OS (hazard ratio [HR]: 0.64 [0.48, 0.87]), PFS (HR: 0.42 [0.34, 0.52]), and CNS-PFS (HR: 0.42 [0.31, 0.57]). The benefits in survival for OS, PFS, and CNS-PFS were validated in nearly all subgroups. Meanwhile, the overall objective response rate (ORR) (risk ratio [RR]: 1.25 [1.02, 1.52]) and CNS-ORR (RR: 1.19 [0.93, 1.51]) also tended to favor the ETC group. However, the addition of chemotherapy also brought about more grade 3-5/serious adverse events (AEs). The top five grade 3-5 AEs in the ETC group were alanine aminotransferase increase (11.25%), neutropenia (7.5%), nausea (7.5%), anorexia (5%), and diarrhea (5%).ConclusionsETC appears to be better than ET in treating EGFR-positive NSCLC patients with BM, with better OS, PFS, CNS-PFS, and responses. However, its poorer safety profile also needs to be taken into consideration.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024551073.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
ObjectiveIn this study, we aimed to perform a network meta-analysis to compare the effectiveness of NAs in decreasing the reactivation of HBV, reducing chemotherapy disruption, and improving survival in oncology patients.MethodsRelevant randomized controlled trials (RCT) evaluating the impact of NAs in HBV infected-related oncology patients were retrieved from electronic databases. The outcome indicators included reactivation rate, survival rate of 1 to 3 years after treatment, and chemotherapy disruption rate. The studies were evaluated for bias using the RCT risk of bias assessment tool recommended in the Cochrane Handbook. The risk ratio (RR) was used to compare the outcome indicators for the anti-viral treatment, and the surface under the cumulative ranking curves (SUCRA) was used to identify the optimal therapeutic regime.ResultsA total of 67 trials containing 5722 patients were included in this study. Regarding the reduction of reactivation rate, entecavir, lamivudine, adefovir alone were less effective than the combination of lamivudine and entecavir (94.9%), with RR values ranging from 3.16 to 3.73. However, based on SUCRA, the efficacy of telbivudine (80.3%) and the combination of lamivudine and adefovir dipivoxil (58.8%) were also acceptable. Entecavir (RR values ranging from 1.25 to 1.50) and lamivudine (RR values ranging from 1.27 to 1.35) can prolong the survival rate of patients at 1-3 years, and were better than adefovir dipivoxil in the comparison of 1-year survival rate. The RR values were 1.18 and 1.19, respectively. And entecavir ‘s ranking in SUCRA was more stable. Entecavir, lamivudine, and tenofovir all reduced chemotherapy interruption rates compared with no antiviral therapy, especially for tenofovir.ConclusionsCurrent evidence shows that lamivudine combined with entecavir, telbivudine, and lamivudine combined with adefovir dipivoxil were the most effective in preventing virus reactivation in HBV infected-related cancer patients treated with chemotherapy. Entecavir had the most stable effect on survival, while tenofovir had the best impact on reducing the chemotherapy disruption rate. Due to limited quality and quantity of the included studies, more high-quality studies are required to verify the above conclusions.Systematic review registrationPROSPEROI [https://www.crd.york.ac.uk/PROSPERO/index.php], identifier CRD4202250685.