Palma Marco
Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain.
Vaccines (Basel). 2025 Mar 24;13(4):344. doi: 10.3390/vaccines13040344.
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body's immune system to precisely target and eliminate cancer cells. However, several key factors influence the selection and effectiveness of these therapies, including BC subtype, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), PD-L1 expression, HER2 resistance, and the tumor microenvironment (TME). BC subtypes play a critical role in shaping treatment responses. Triple-negative breast cancer (TNBC) exhibits the highest sensitivity to immunotherapy, while HER2-positive and hormone receptor-positive (HR+) subtypes often require combination strategies for optimal outcomes. High TMB enhances immune responses by generating neoantigens, making tumors more susceptible to immune checkpoint inhibitors (ICIs); whereas, low TMB may indicate resistance. Similarly, elevated TIL levels are associated with better immunotherapy efficacy, while PD-L1 expression serves as a key predictor of checkpoint inhibitor success. Meanwhile, HER2 resistance and an immunosuppressive TME contribute to immune evasion, highlighting the need for multi-faceted treatment approaches. Current breast cancer immunotherapies encompass a range of targeted treatments. HER2-directed therapies, such as trastuzumab and pertuzumab, block HER2 dimerization and enhance antibody-dependent cellular cytotoxicity (ADCC), while small-molecule inhibitors, like lapatinib and tucatinib, suppress HER2 signaling to curb tumor growth. Antibody-drug conjugates (ADCs) improve tumor targeting by coupling monoclonal antibodies with cytotoxic agents, minimizing off-target effects. Meanwhile, ICIs, including pembrolizumab, restore T-cell function, and CAR-macrophage (CAR-M) therapy leverages macrophages to reshape the TME and overcome immunotherapy resistance. While immunotherapy, particularly in TNBC, has demonstrated promise by eliciting durable immune responses, its efficacy varies across subtypes. Challenges such as immune-related adverse events, resistance mechanisms, high costs, and delayed responses remain barriers to widespread success. Breast cancer vaccines-including protein-based, whole-cell, mRNA, dendritic cell, and epitope-based vaccines-aim to stimulate tumor-specific immunity. Though clinical success has been limited, ongoing research is refining vaccine formulations, integrating combination therapies, and identifying biomarkers for improved patient stratification. Future advancements in BC treatment will depend on optimizing immunotherapy through biomarker-driven approaches, addressing tumor heterogeneity, and developing innovative combination therapies to overcome resistance. By leveraging these strategies, researchers aim to enhance treatment efficacy and ultimately improve patient outcomes.
乳腺癌(BC)因其复杂的生物学特性,仍是一项重大的全球健康挑战,这使得诊断和治疗都变得复杂。免疫疗法和癌症疫苗已成为有前景的替代方案,利用人体免疫系统精准靶向并消除癌细胞。然而,有几个关键因素会影响这些疗法的选择和效果,包括BC亚型、肿瘤突变负荷(TMB)、肿瘤浸润淋巴细胞(TILs)、PD-L1表达、HER2耐药性以及肿瘤微环境(TME)。BC亚型在塑造治疗反应方面起着关键作用。三阴性乳腺癌(TNBC)对免疫疗法表现出最高的敏感性,而HER2阳性和激素受体阳性(HR+)亚型通常需要联合策略以获得最佳疗效。高TMB通过产生新抗原增强免疫反应,使肿瘤更容易受到免疫检查点抑制剂(ICI)的影响;而低TMB可能表明存在耐药性。同样,TIL水平升高与更好的免疫治疗疗效相关,而PD-L1表达是检查点抑制剂成功的关键预测指标。同时,HER2耐药性和免疫抑制性TME导致免疫逃逸,凸显了多方面治疗方法的必要性。目前的乳腺癌免疫疗法包括一系列靶向治疗。针对HER2的疗法,如曲妥珠单抗和帕妥珠单抗,可阻断HER2二聚化并增强抗体依赖性细胞毒性(ADCC),而小分子抑制剂,如拉帕替尼和图卡替尼,可抑制HER2信号传导以抑制肿瘤生长。抗体药物偶联物(ADC)通过将单克隆抗体与细胞毒性药物偶联来改善肿瘤靶向性,将脱靶效应降至最低。同时,包括派姆单抗在内的ICI可恢复T细胞功能,而嵌合抗原受体巨噬细胞(CAR-M)疗法利用巨噬细胞重塑TME并克服免疫治疗耐药性。虽然免疫疗法,特别是在TNBC中,通过引发持久的免疫反应已显示出前景,但其疗效在不同亚型中有所不同。免疫相关不良事件、耐药机制、高成本和反应延迟等挑战仍然是广泛成功的障碍。乳腺癌疫苗——包括基于蛋白质的、全细胞的、mRNA的、树突状细胞的和基于表位的疫苗——旨在刺激肿瘤特异性免疫。尽管临床成功有限,但正在进行的研究正在完善疫苗配方、整合联合疗法并确定生物标志物以改善患者分层。BC治疗的未来进展将取决于通过生物标志物驱动的方法优化免疫疗法、解决肿瘤异质性以及开发创新的联合疗法以克服耐药性。通过利用这些策略,研究人员旨在提高治疗效果并最终改善患者预后。