Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Jul 24, 2025; 16(7): 104959
Published online Jul 24, 2025. doi: 10.5306/wjco.v16.i7.104959
Expanding horizons in esophageal squamous cell carcinoma: The promise of induction chemoimmunotherapy with radiotherapy
Wenxue Ma, Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, San Diego, CA 92093, United States
Natalia Baran, Department of Hematology and Central Hematology Laboratory, Bern University Hospital, University of Bern, Bern 3012, Switzerland
Natalia Baran, Institute of Hematology and Transfusion Medicine, Section of Experimental Hematology, Warsaw, 02-776, Poland
ORCID number: Wenxue Ma (0000-0001-9228-6162); Natalia Baran (0000-0003-0618-4798).
Author contributions: Ma W reviewed the relevant literature, conceptualized and outlined this editorial, wrote, edited, and finalized the manuscript for publication; Baran N reviewed the manuscript and provided editorial feedback and revisions.
Supported by National Science Centre, Poland, No. 2021/43/B/NZ5/03345.
Conflict-of-interest statement: The authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Wenxue Ma, MD, PhD, Professor, Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, 2880 Torrey Pines Scenic Drive, San Diego, CA 92093, United States. wma@health.ucsd.edu
Received: January 7, 2025
Revised: February 23, 2025
Accepted: March 13, 2025
Published online: July 24, 2025
Processing time: 196 Days and 16.4 Hours

Abstract

Esophageal squamous cell carcinoma (ESCC) remains a highly aggressive malignancy with limited effective therapeutic options for patients with locally advanced unresectable disease. The study by Wei et al, featured in this issue, highlights the potential of induction chemoimmunotherapy followed by definitive radiotherapy or concurrent chemoradiotherapy to improve treatment outcomes in this challenging patient population. This retrospective analysis of 132 patients demonstrates promising results, including a median progression-free survival of 14.2 months and overall survival of 19.9 months, alongside an acceptable safety profile. Notably, the study identifies the effectiveness of induction therapy and maintenance immunotherapy as key prognostic factors, emphasizing the synergistic potential of integrating immune checkpoint inhibitors with radiotherapy. While these findings are encouraging, they require further validation through prospective trials, along with biomarker-based and immune response studies, to refine patient selection and maximize therapeutic benefits. This editorial explores the implications of this research, its impact on clinical practice, and future directions for advancing the treatment landscape of ESCC.

Key Words: Esophageal squamous cell carcinoma; Chemoimmunotherapy, Radiotherapy; Immune checkpoint inhibitors; Locally advanced cancer; Treatment outcomes

Core Tip: This editorial highlights the study by Wei et al, which examines the impact of induction chemoimmunotherapy followed by radiotherapy or concurrent chemoradiotherapy in patients with locally advanced esophageal squamous cell carcinoma (ESCC). The study demonstrates improved survival outcomes and a manageable safety profile, identifies the effectiveness of induction therapy and maintenance immunotherapy as key prognostic factors. This innovative approach represents a promising advancement in the treatment of ESCC and underscores the need for future prospective trials and mechanistic studies incorporating biomarker investigations to refine therapeutic strategies and enhance patient outcomes.



INTRODUCTION

Cancer remains a major global health challenge, with an estimated 19.3 million new cases and 10 million cancer-related deaths worldwide in 2022[1]. Advances in cancer treatment, ranging from traditional approaches to emerging therapies, have been crucial in improving patient outcomes[2]. Esophageal cancer ranks among the most lethal malignancies, with 510716 new cases and 445129 deaths, making it the 11th most common cancer and the 7th leading cause of cancer-related mortality globally[1]. Despite advancements in diagnosis and treatment, the overall five-year survival rate for esophageal cancer remains dismal at approximately 20%[3].

Among the histological subtypes, esophageal squamous cell carcinoma (ESCC) poses a significant challenge, particularly in China, where it accounts for the majority of esophageal cancer cases[4]. Patients with locally advanced unresectable ESCC face especially poor prognoses, with five-year survival rates ranging from 10% to 30% or even lower, reflecting both global trends and regional variations influenced by factors such as healthcare access, treatment availability, and early detection rates[5,6]. These alarming statistics underscore the urgent need for improved therapeutic strategies and prognostic biomarkers to guide clinical decision-making and enhance patient outcomes[2,7].

Over the past decades, cancer therapies have evolved significantly, transitioning from surgery and radiotherapy to the advent of chemotherapy, targeted therapies, and immunotherapy[1]. Despite these advancements, the standard treatment for locally advanced ESCC remains chemoradiotherapy[5], which is an effective regimen yet often associated with high toxicity and limited long-term survival benefits[8]. Furthermore, recurrence rates remain high, and not all patients respond favorably to existing regimens[9]. This highlights a critical gap in the treatment landscape, where durable tumor control and improved survival outcomes remain elusive for many patients[10]. Addressing these unmet needs requires innovative treatment combinations that enhance efficacy while maintaining tolerability[11].

The recent focus on combining treatment modalities, such as chemoimmunotherapy with radiotherapy, has emerged as a promising approach to improve tumor control and survival while managing toxicity[12]. Such strategies aim to exploit the synergistic effects of chemotherapy, immunotherapy, and radiotherapy[13]. In ESCC, this multimodal combination represents a novel therapeutic avenue that could potentially overcome the limitations of conventional treatments, offering a pathway to more durable responses and prolonged survival[14].

Treating ESCC with multimodal approaches is challenging due to several factors. Anatomically, the proximity of the esophagus to vital structures such as trachea, heart, and major blood vessels limits surgical options and the safe delivery of radiotherapy. Clinically, ESCC is often diagnosed at advanced stages because early symptoms are nonspecific, reducing the effectiveness of localized treatments[15]. Biologically, the tumor’s heterogeneity and its tendency to spread through lymphatic channels complicate disease control[16]. Although chemoradiotherapy is the standard treatment, it often causes significant side effects, such as esophagitis and nutritional issues, which can limit patient tolerance to therapy[17]. Additionally, the tumor microenvironment (TME) may further suppress immune responses, reducing the effectiveness of immunotherapies[18,19]. Patient-related factors, including malnutrition, comorbidities, and poor overall health, may also preclude aggressive treatment options. These challenges underscore the urgent need for personalized approaches that balance treatment efficacy with patient safety.

In this issue of the World Journal of Clinical Oncology, the study by Wei et al[20] critically examines the use of induction chemoimmunotherapy followed by definitive radiotherapy or concurrent chemoradiotherapy as a promising strategy to address this critical unmet clinical need. By incorporating immune checkpoint inhibitors (ICIs) with standard chemotherapy and radiotherapy protocols, this approach seeks to improve treatment response rates, prolong progression-free survival (PFS), and enhance overall survival (OS) while mitigating the adverse effects commonly associated with conventional therapies[20,21]. The findings of this study have the potential to reshape current treatment paradigms and offer new hope for patients with advanced ESCC, who have historically faced limited therapeutic options and poor prognoses.

KEY INSIGHTS AND INNOVATIONS

This retrospective study offers a comprehensive evaluation of combining chemotherapy with ICIs and radiotherapy in patients with locally advanced ESCC[22,23]. The findings are particularly noteworthy when compared to conventional chemoradiotherapy (CRT) approaches.

In terms of safety profile, treatment-related adverse events (AEs) aligned with expectations, with no treatment-related deaths[24]. Grade 3 and 4 toxicities were common, with esophagitis occurring in 63.6% of patients and neutropenia in 25%. Postoperative Grade 3-4 morbidities were less frequent, including surgical wound infection (2.3%), acute renal failure (2.3%), and anastomotic stricture (2.3%). These rates are comparable to those reported in previous studies evaluating chemoradiotherapy alone, where grade 3 or higher toxicities have ranged from 60% to 70% for esophagitis and approximately 20% to 30% for neutropenia[25]. Importantly, the addition of ICIs did not significantly increase the incidence of severe AEs compared to historical data, underscoring the manageable safety profile of this regimen despite incorporating immunotherapy.

For survival outcomes, the study reported a median PFS of 14.2 months and OS of 19.9 months, which represent significant improvements over historical benchmarks with standard CRT, where median PFS typically ranges from 8 to 10 months, and median OS from 12 to 16 months[26]. A similar study reported a median PFS of 9.7 months and an OS of 18.5 months in patients undergoing definitive chemoradiotherapy, further supporting these survival benchmarks[27]. This improvement highlights the potential survival benefit of integrating induction chemoimmunotherapy with radiotherapy compared to traditional approaches. Additionally, the study identified the effectiveness of induction therapy and maintenance immunotherapy as independent prognostic factors for OS, highlighting their critical role in improving patient outcomes[28].

These findings are particularly significant given the challenges associated with combining systemic therapies and radiotherapy in patients who have historically faced poor prognoses, high toxicity risks, and limited treatment options. By demonstrating improved survival without a corresponding increase in severe AEs, this approach offers a clinically meaningful advancement over conventional CRT regimens.

CLINICAL AND RESEARCH IMPLICATIONS

The study by Wei et al[20] highlights the therapeutic potential of integrating ICIs, including pembrolizumab and camrelizumab, with conventional therapies, underscoring the synergy between immunotherapy and radiotherapy. This combination appears to enhance tumor control while maintaining an acceptable safety profile[22,29]. However, the translation of these findings into clinical practice presents several practical challenges, warranting careful consideration. While the combination of immunotherapy and radiotherapy shows promising efficacy, understanding the full spectrum of treatment-related AEs, including long-term toxicities, remains essential[30]. Potential long-term toxicities of this combination therapy include radiation-induced esophageal strictures, pulmonary fibrosis, cardiotoxicity, and chronic dysphagia[31], all of which may significantly impact patients' quality of life. Additionally, ICIs have been associated with prolonged immune-related AEs (irAEs), such as endocrine dysfunction (e.g., hypothyroidism, adrenal insufficiency), persistent pneumonitis, and chronic colitis[32,33]. Given these risks, long-term follow-up and proactive management strategies are crucial to optimizing patient outcomes while minimizing late-onset complications[22]. Future studies should prioritize comprehensive safety assessments and strategies to manage potential side effects in diverse patient populations.

The TME, including immune evasion pathways such as CD47-SIRPα signaling, plays a pivotal role in determining treatment response[34], particularly in ESCC[35]. The TME comprises immune cells, stromal cells, blood vessels, and extracellular matrix components, all orchestrated by cytokines and soluble mediators that modulate tumor cell behavior, disease progression and therapy outcomes[18,36]. An immunosuppressive TME, often driven by CD39-mediated adenosine signaling, characterized by high levels of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages[37,38], can hinder the efficacy of both radiotherapy and ICIs[39].

Radiotherapy has been shown to modulate the TME through multiple mechanisms[40]. On one hand, it can enhance antitumor immunity by promoting antigen release, increasing major histocompatibility complex expression, and facilitating T-cell infiltration into tumors[41]. On the other hand, radiotherapy may also induce immunosuppressive effects that limit sustained immune responses[42]. These effects include the upregulation of programmed death-ligand 1 (PD-L1) on tumor and immune cells, increasing recruitment of Tregs and MDSCs[43], and the release of immunosuppressive cytokines such as TGF-β and IL-10[44]. Additionally, high-dose radiation can damage vascular integrity within the tumor, reducing immune cell infiltration[40], while low-dose radiation may reprogram certain immune cells into more suppressive phenotypes[45]. The dual nature of radiotherapy underscores the complexity of its interaction with the immune system.

Despite these promising mechanistic insights, several practical challenges arise when translating combination therapies into routine clinical practice. Managing treatment-related AEs, such as esophagitis, hematologic toxicities, and irAEs, is critical to preventing treatment interruptions and ensuring patient adherence[33,46]. Management strategies, including prophylactic supportive care and dose modifications, play a key role in mitigating these toxicities and improving patient outcomes[47]. Early initiation of supportive measures (e.g., nutritional support, growth factor administration, and corticosteroids for irAEs) is essential[48]. Patient-specific factors, including comorbidities, age, and performance status, may limit tolerance to aggressive regimens, underscoring the importance of individualized treatment planning[49]. Real-world considerations, such as resource availability, cost, and patient compliance with frequent treatment visits, can also impact therapy implementation. Multidisciplinary care teams and clear patient education are vital in addressing these challenges. Future research should explore adaptive treatment strategies, such as precision dosing using model-informed reinforcement learning, to improve tolerability without compromising efficacy[50]. By addressing these practical concerns, clinicians can optimize patient outcomes while minimizing treatment-related risks.

MECHANISMS OF ICI AND RADIOTHERAPY SYNERGY

Combining ICIs with radiotherapy aims to overcome these barriers by enhancing antitumor immunity while counteracting TME-mediated resistance[40,51]. Radiotherapy-induced tumor cell death promotes the release of tumor-associated antigens, facilitating antigen presentation by dendritic cells and triggering the activation of cytotoxic CD8+ T cells[40,52]. However, sustained antigen exposure can lead to T-cell exhaustion, characterized by the upregulation of inhibitory receptors such as programmed cell death protein 1 (PD-1), lymphocyte activation gene 3 (LAG-3), and TIM-3, which impairs T-cell functionality[53,54]. Strategies aimed at reversing T-cell exhaustion, such as mitochondrial modulation as well as combination therapies targeting multiple immune checkpoints and cytokine signaling have shown promise in restoring anti-tumor immunity[55]. For example, dual blockade of PD-1 and LAG-3 has been demonstrated to enhance T-cell reinvigoration, while cytokine-based interventions, including IL-2 and IL-12, have been explored to promote effector T-cell function and improve response durability in ESCC[56]. ICIs, particularly those targeting the PD-1/PD-L1 axis, help to reverse T-cell exhaustion, restore effector T-cell activity, thereby enhancing antitumor responses[57].

In addition to modulating T-cell responses, the cytokine milieu plays a crucial role in shaping treatment outcomes. Radiotherapy can induce the release of pro-inflammatory cytokines, such as interferon-gamma (IFN-γ) and IL-2, which promote immune activation and T-cell recruitment[58]. However, these effects are often counterbalanced by the simultaneous release of immunosuppressive cytokines like TGF-β and IL-10, which inhibit effective antitumor immunity[40]. Targeting these cytokine pathways in combination with ICIs and radiotherapy may further enhance immune activation and improve treatment efficacy[59].

Additionally, immune activation pathways involving STING signaling and cGAS-STING pathways are activated following radiotherapy-induced DNA damage, leading to the production of type I interferons (IFNs) that promote dendritic cell maturation and T-cell priming[60]. The integration of ICIs into this context amplifies the T-cell-mediated cytotoxic response, creating a robust immune attack against the tumor[61]. Emerging strategies, such as combining radiotherapy with STING agonists or targeting MDSCs that contribute to immune evasion, are being explored to further optimize the efficacy of this multimodal approach[62].

CLINICAL IMPLICATIONS AND FUTURE DIRECTIONS

Understanding and targeting the TME, cytokine release patterns, T-cell exhaustion pathways, and immune activation cascades are crucial for optimizing therapeutic strategies and improving patient outcomes in ESCC[63]. However, several questions remain that warrant further investigation.

First, the interplay between radiotherapy and the immune microenvironment, including potential immunosuppressive effects, requires deeper exploration[64]. Second, identifying biomarkers predictive of treatment responses is critical for enhancing the clinical utility of this regimen[65]. Ongoing efforts focus on evaluating PD-L1 expression, which has been widely studied as a potential predictor of response to ICIs[66]; however, its predictive value in ESCC remains inconsistent due to variability in detection methods and cutoff thresholds[67]. Another biomarker is circulating tumor DNA (ctDNA) has emerged as a promising non-invasive biomarker, enabling the monitoring of tumor burden, detection of minimal residual disease, and prediction of treatment response, thereby supporting personalized oncology approaches[68]. Furthermore, dynamic changes in ctDNA levels during therapy have been associated with survival outcomes and could serve as an early indicator of treatment efficacy[69]. Despite these advancements, challenges remain, including standardizing testing protocols, establishing universally accepted thresholds, and integrating biomarker data into clinical decision-making[70]. Further research is needed to validate these biomarkers in larger, prospective cohorts.

Finally, the retrospective design of this study and its limited sample size necessitate validation through well-designed prospective trials[71,72]. Such prospective studies are essential for minimizing bias, enhancing generalizability, and providing more robust evidence of the treatment’s efficacy and safety in diverse patient populations.

LIMITATIONS AND FUTURE RESEARCH NEEDS

While the results of this study are promising, several limitations should be acknowledged. First, the retrospective study design is inherently prone to selection bias, information bias, and confounding factors that may affect the generalizability of the findings[73]. Second, the limited sample size restricts the statistical power of the study and may not adequately capture the variability in patient responses or rare AEs[74]. This limitation highlights the necessity of conducting power analyses in future studies to determine an adequate sample size capable of detecting clinically meaningful differences with statistical significance[75].

To address these limitations, there is an urgent need for well-designed, prospective multicenter clinical trials to validate the efficacy, safety, and long-term outcomes of the combination therapy across diverse patient populations[76]. Such trials would not only enhance the robustness of the findings but also improve their generalizability to different clinical settings. As emphasized by Subbiah[77], global collaboration in multicenter studies is essential to account for regional variations in treatment response, healthcare access, and genetic diversity, ultimately ensuring broader applicability of results.

BIOMARKERS AND PERSONALIZED TREATMENT STRATEGIES IN CLINICAL PRACTICE

Biomarkers play a pivotal role in predicting treatment responses, monitoring therapeutic efficacy, and guiding personalized treatment strategies in ESCC[78]. Despite the promising results of combining ICIs with radiotherapy, heterogeneous patient responses underscore the need for reliable predictive biomarkers to improve patient selection and outcomes[33,79].

Among the most widely studied biomarkers is PD-L1 expression, which is often used to predict ICI responsiveness[67,80]. Higher PD-L1 levels correlate with improved outcomes in some studies; however, its predictive value remains inconsistent due to variations in detection methods, cutoff thresholds, and tumor heterogeneity[69]. Ongoing trials are working to establish standardized PD-L1 scoring systems to enhance predictive accuracy[69,81].

Another promising biomarker is ctDNA, which provides a non-invasive approach for tracking tumor burden, detecting minimal residual disease, and predicting early treatment response[82]. Fluctuations in ctDNA levels during therapy have been linked to PFS and OS[69]. Active clinical trials aim to validate ctDNA-guided treatment adjustments in ESCC[82,83].

Additional biomarkers, such as tumor mutational burden (TMB) and microsatellite instability (MSI), have shown potential in predicting ICI efficacy, with higher TMB and MSI-high tumors generally associated with better responses[84]. While these markers are well established in other cancers, their clinical relevance in ESCC requires further investigation[85]. Advances in next-generation sequencing (NGS) and multi-analyte platforms, including liquid biopsy technologies are enabling the development of multi-biomarker signatures such as PD-L1, TMB, MSI, and immune gene profiles for improved patient stratification and early detection[86].

Beyond molecular biomarkers, personalized treatment strategies should also consider TME characteristics and patient-specific factors[36]. Tumors with immune-inflamed phenotypes, characterized by high CD8+ T-cell infiltration and elevated IFN-γ signatures, are more likely to benefit from ICI-based therapies[87,88]. Conversely, immune-excluded or immune-desert phenotypes may require strategies to modulate the TME, such as combining radiotherapy to enhance immune cell infiltration[89].

NGS-based genetic profiling can identify actionable mutations, enabling tailored therapy combinations[90]. For instance, patients with high TMB or specific immune gene expression profiles may benefit from intensified combination regimens, while those with low biomarker expression may be considered for alternative approaches[91]. Emerging evidence suggests that dynamic biomarker monitoring (e.g., serial ctDNA measurements) can help adjust treatment plans in real-time, further personalizing therapy[92].

In clinical practice, integrating biomarker data into treatment algorithms ensures that patients most likely to benefit from combination therapies receive them while minimizing unnecessary toxicity in non-responders[93]. Prospective, biomarker-driven trials are essential to establish the clinical utility of these markers and to develop personalized decision-making frameworks for ESCC[94]. Such trials should also include diverse patient populations, accounting for variations in age, gender, comorbidities, and tumor staging to enhance the generalizability of findings[95].

FUTURE DIRECTIONS

The findings presented in this study pave the way for significant advancements in management of ESCC[96]. Incorporating maintenance immunotherapy into treatment regimens holds the potential to achieve sustained tumor control[97,98]. Additionally, exploring dose escalation strategies and implementing novel radiation techniques, such as proton therapy and intensity-modulated proton therapy, could further enhance treatment outcomes by delivering higher radiation doses to tumors while sparing surrounding healthy tissues, thereby reducing toxicity and improving patient tolerance[99]. Both adaptive radiotherapy, which allows for real-time adjustments based on patient-specific anatomical and tumor changes, and proton-based therapies, known for their favorable dose distribution, represent promising approaches to maximize therapeutic efficacy while minimizing collateral damage to normal tissues[100,101]. Future clinical studies should consider integrating these technologies to further refine treatment protocols and optimize patient outcomes.

Beyond clinical application, investigating multimodal treatment strategies that extend beyond the combination of ICIs and radiotherapy is crucial for enhancing therapeutic efficacy[102]. Future research should explore the potential synergistic effects of combining ICIs with targeted therapies, chemotherapy, and other modalities to overcome resistance and improve patient outcomes[103]. Such approaches may offer distinct advantages in different molecular subtypes and stages of esophageal cancer, underscoring the need for personalized treatment strategies[104]. Key areas of interest include identifying optimal sequencing and dosing regimens, as well as understanding how these combinations affect tumor biology, immune modulation, and treatment resistance[105].

Preclinical research into the mechanisms underlying the synergy between ICIs and radiotherapy will be essential for refining and expanding these therapeutic approaches[51]. Future studies should focus on elucidating the biological pathways that contribute to overcoming immunotherapy resistance and enhancing radiotherapy response[41]. Key areas of interest include understanding how radiotherapy modulates the TME, enhances antigen presentation, and promotes immune cell infiltration[106]. Investigating the role of the cGAS-STING pathway activation, changes in MDSCs, and the influence of Tregs following combined therapy could provide critical insights into mechanisms of action and resistance[107].

To facilitate these mechanistic studies, patient-derived xenograft (PDX) and syngeneic mouse models are valuable preclinical tools. PDX models, which involve implanting human tumor tissues into immunodeficient mice, can recapitulate the complexity and heterogeneity of human tumors, allowing for the assessment of therapeutic efficacy and resistance mechanisms in a patient-relevant context[108]. Syngeneic models, which use immunocompetent mice bearing murine tumors, are particularly useful for studying immune-tumor interactions and the immune-mediated effects of combination therapies such as ICIs and radiotherapy[109]. Incorporating these models into future studies could elucidate the contribution of different immune cell populations to treatment outcomes and help identify predictive biomarkers, ultimately guiding the development of more effective ESCC treatment strategies[110].

Identifying mechanisms of drug resistance remains a significant barrier to long-term treatment efficacy. Techniques such as CRISPR-Cas9 screening offer powerful tools for discovering key genetic drivers of resistance to chemoimmunotherapy and radiotherapy[111]. For instance, recent studies have demonstrated how genome-wide loss-of-function screens can uncover critical resistance pathways, providing potential targets for combination therapies[112]. Integrating such approaches into ESCC research could facilitate the development of strategies to overcome resistance, personalize treatment regimens, and ultimately improve patient outcomes[113]. Moreover, incorporating molecular profiling into clinical trials could enhance patient stratification, enabling biomarker-driven treatment decisions and better identification of patients most likely to benefit from multimodal therapies[114].

Addressing patient diversity in future studies is equally crucial for enhancing the applicability and generalizability of research findings[115]. The current study, while informative, is limited by its sample size and may not fully capture the variability in clinical characteristics such as age, gender, comorbidities, and tumor staging[116]. Future prospective multicenter trials should prioritize the inclusion of diverse patient populations to better reflect real-world clinical settings and ensure broader applicability[117]. Incorporating patients from different demographic backgrounds and with varied clinical profiles will provide more comprehensive insights into treatment efficacy, safety, and patient-specific outcomes, ultimately improving clinical decision-making and patient care[118].

Lastly, evaluating the long-term effects of combining immunotherapy with radiotherapy is vital. Long-term follow-up studies are essential to assess the durability of treatment benefits, monitor potential late-onset toxicities, and understand the impact on OS and quality of life[119]. Such data will be critical in optimizing treatment regimens for sustained patient benefit while minimizing adverse effects.

CONCLUSION

The study by Wei et al[20] represents a significant advancement in addressing the poor prognosis associated with advanced ESCC[120]. By integrating induction chemoimmunotherapy with radiotherapy in a clinically well-characterized ESCC cohort, this research provides valuable insights into treatment efficacy and safety. The findings highlight the potential of combining established therapies with innovative approaches to expand therapeutic options and improve patient outcomes[121].

To fully validate these findings and optimize treatment strategies, there is an urgent need for well-designed, multicenter, prospective trials to assess the efficacy, safety, and long-term outcomes of combination therapies across diverse patient populations[122]. Furthermore, collaborative efforts at a global scale will be essential for translating these promising results into standardized clinical practice[123,124]. As the field progresses, such insights will play a crucial role in reshaping the prognosis and outcomes for patients with this challenging malignancy.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: United States

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade B

Novelty: Grade A, Grade A, Grade B

Creativity or Innovation: Grade A, Grade A, Grade B

Scientific Significance: Grade A, Grade B, Grade B

P-Reviewer: Zhang L; Xu WS S-Editor: Qu XL L-Editor: A P-Editor: Zhao YQ

References
1.  Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5690]  [Cited by in RCA: 7675]  [Article Influence: 7675.0]  [Reference Citation Analysis (2)]
2.  Sonkin D, Thomas A, Teicher BA. Cancer treatments: Past, present, and future. Cancer Genet. 2024;286-287:18-24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 160]  [Article Influence: 160.0]  [Reference Citation Analysis (0)]
3.  Godefa TM, Derks S, Thijssen VLJL. Galectins in Esophageal Cancer: Current Knowledge and Future Perspectives. Cancers (Basel). 2022;14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
4.  Zhu H, Ma X, Ye T, Wang H, Wang Z, Liu Q, Zhao K. Esophageal cancer in China: Practice and research in the new era. Int J Cancer. 2023;152:1741-1751.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 93]  [Article Influence: 46.5]  [Reference Citation Analysis (0)]
5.  Xie R, Cai Q, Chen T, Huang H, Chen C. Current and future on definitive concurrent chemoradiotherapy for inoperable locally advanced esophageal squamous cell carcinoma. Front Oncol. 2024;14:1303068.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
6.  DaSilva LL, Aguiar PN Jr, de Lima Lopes G. Immunotherapy for Advanced Esophageal Squamous Cell Carcinoma-Renewed Enthusiasm and a Lingering Challenge. JAMA Oncol. 2021;7:1613-1614.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
7.  Christodoulidis G, Agko SE, Koumarelas KE, Kouliou MN, Zacharoulis D. Advancements and challenges in the treatment of esophageal cancer: A comprehensive review. World J Clin Oncol. 2024;15:1463-1467.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
8.  Wang X, Han W, Zhang W, Wang X, Ge X, Lin Y, Zhou H, Hu M, Wang W, Liu K, Lu J, Qie S, Zhang J, Deng W, Wang L, Han C, Li M, Zhang K, Li L, Wang Q, Shi H, Yu Z, Zhao Y, Sun X, Shi Y, Pang Q, Zhou Z, Liang J, Chen D, Feng Q, Bi N, Zhang T, Deng L, Wang W, Liu W, Wang J, Zhai Y, Wang J, Chen W, Chen J, Xiao Z; Jing-Jin-Ji Esophageal and Esophagogastric Cancer Radiotherapy Oncology Group (3JECROG). Effectiveness of S-1-Based Chemoradiotherapy in Patients 70 Years and Older With Esophageal Squamous Cell Carcinoma: A Randomized Clinical Trial. JAMA Netw Open. 2023;6:e2312625.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 15]  [Cited by in RCA: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
9.  Yang Z, Tian H, Chen X, Li B, Bai G, Cai Q, Xu J, Guo W, Wang S, Peng Y, Liang Q, Xue L, Gao S. Single-cell sequencing reveals immune features of treatment response to neoadjuvant immunochemotherapy in esophageal squamous cell carcinoma. Nat Commun. 2024;15:9097.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
10.  Elmore LW, Greer SF, Daniels EC, Saxe CC, Melner MH, Krawiec GM, Cance WG, Phelps WC. Blueprint for cancer research: Critical gaps and opportunities. CA Cancer J Clin. 2021;71:107-139.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 62]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
11.  Jabbour SK, Kumar R, Anderson B, Chino JP, Jethwa KR, McDowell L, Lo AC, Owen D, Pollom EL, Tree AC, Tsang DS, Yom SS. Combinatorial Approaches for Chemotherapies and Targeted Therapies With Radiation: United Efforts to Innovate in Patient Care. Int J Radiat Oncol Biol Phys. 2024;118:1240-1261.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
12.  Higuchi T, Shoji Y, Koyanagi K, Tajima K, Kanamori K, Ogimi M, Yatabe K, Ninomiya Y, Yamamoto M, Kazuno A, Nabeshima K, Nakamura K. Multimodal Treatment Strategies to Improve the Prognosis of Locally Advanced Thoracic Esophageal Squamous Cell Carcinoma: A Narrative Review. Cancers (Basel). 2022;15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
13.  Gao Z, Zhao Q, Xu Y, Wang L. Improving the efficacy of combined radiotherapy and immunotherapy: focusing on the effects of radiosensitivity. Radiat Oncol. 2023;18:89.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
14.  Ooki A, Osumi H, Chin K, Watanabe M, Yamaguchi K. Potent molecular-targeted therapies for advanced esophageal squamous cell carcinoma. Ther Adv Med Oncol. 2023;15:17588359221138377.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
15.  Deboever N, Jones CM, Yamashita K, Ajani JA, Hofstetter WL. Advances in diagnosis and management of cancer of the esophagus. BMJ. 2024;385:e074962.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 44]  [Reference Citation Analysis (0)]
16.  Guo D, Sheng K, Zhang Q, Li P, Sun H, Wang Y, Lyu X, Jia Y, Wang C, Wu J, Zhang X, Wang D, Sun Y, Huang S, Yu J, Zhang J. Single-cell transcriptomic analysis reveals the landscape of epithelial-mesenchymal transition molecular heterogeneity in esophageal squamous cell carcinoma. Cancer Lett. 2024;587:216723.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 10]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
17.  Kariya M, Yamamoto K, Kawamura A, Tanizaki S, Ueda K, Sai K, Hata A. Cystine and theanine for chemoradiotherapy-induced esophagitis in non-small cell lung cancer: a prospective observational study. Support Care Cancer. 2024;32:400.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
18.  Xu S, Li X, Ma W. Redefining the tumor microenvironment with emerging therapeutic strategies. Oncol Res. 2024;32:1701-1708.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
19.  Czajka-Francuz P, Prendes MJ, Mankan A, Quintana Á, Pabla S, Ramkissoon S, Jensen TJ, Peiró S, Severson EA, Achyut BR, Vidal L, Poelman M, Saini KS. Mechanisms of immune modulation in the tumor microenvironment and implications for targeted therapy. Front Oncol. 2023;13:1200646.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
20.  Wei Z, Wang L, Wang RQ, Wang Y, Chen H, Ma HL, Xu YJ. Safety and Effectiveness of Induction Chemoimmunotherapy Followed by Definitive Radiotherapy or Concurrent Chemoradiotherapy in Esophageal Squamous Cell Carcinoma. World J Clin Oncol. 2025;16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
21.  Wei J, Li W, Zhang P, Guo F, Liu M. Current trends in sensitizing immune checkpoint inhibitors for cancer treatment. Mol Cancer. 2024;23:279.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
22.  Yap DWT, Leone AG, Wong NZH, Zhao JJ, Tey JCS, Sundar R, Pietrantonio F. Effectiveness of Immune Checkpoint Inhibitors in Patients With Advanced Esophageal Squamous Cell Carcinoma: A Meta-analysis Including Low PD-L1 Subgroups. JAMA Oncol. 2023;9:215-224.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 15]  [Cited by in RCA: 48]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
23.  Jiang M, Hu Y, Lin G, Chen C, Li H. Radiotherapy combined with immune checkpoint inhibitors in locally advanced/metastatic esophageal squamous cell carcinoma: clinical trials, efficacy and future directions. Front Immunol. 2023;14:1177085.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
24.  Guo JC, Huang TC, Kuo HY, Lin CC, Hsu FM, Cheng JC, Huang YL, Hsieh MS, Huang PM, Lee JM, Wu SL, Hsu CH. Adjuvant chemoradiotherapy plus pembrolizumab for locally advanced esophageal squamous cell carcinoma with high risk of recurrence following neoadjuvant chemoradiotherapy: a single-arm phase II study. Cancer Immunol Immunother. 2024;73:230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
25.  Chen M, Huang Y, Zhang S, Zeng T, Huang G, Chen C, Zheng B. Safety and efficacy of camrelizumab combined with radiotherapy as neoadjuvant therapy for locally advanced esophageal squamous cell carcinoma: a prospective single-arm phase II clinical trial protocol. Trials. 2023;24:554.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
26.  Lertbutsayanukul C, Tharavej C, Klaikeaw N, Prayongrat A, Lowanitchai C, Sriuranpong V. High dose radiation with chemotherapy followed by salvage esophagectomy among patients with locally advanced esophageal squamous cell carcinoma. Thorac Cancer. 2017;8:219-228.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
27.  Shi Z, Zhu X, Ruan C, Wei G, Li J, Qiu H, Gao L, Cai G, Zhangcai Y, Li B, Wang J, Gong Y, Chen J, Zhao W, Wu Y, Ke S, Chen Y. Evaluation of Concurrent Chemoradiotherapy for Survival Outcomes in Patients With Synchronous Oligometastatic Esophageal Squamous Cell Carcinoma. JAMA Netw Open. 2022;5:e2244619.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
28.  Lian HM, Wu JL, Liufu WJ, Yu TT, Niu SQ, Bao Y, Peng F. Induction immunotherapy plus chemotherapy followed by definitive chemoradiation therapy in locally advanced esophageal squamous cell carcinoma: a propensity-score matched study. Cancer Immunol Immunother. 2024;73:55.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
29.  Gao Z, Huang S, Wang S, Tang D, Xu W, Zeng R, Qiao G. Efficacy and safety of immunochemotherapy, immunotherapy, chemotherapy, and targeted therapy as first-line treatment for advanced and metastatic esophageal cancer: a systematic review and network meta-analysis. Lancet Reg Health West Pac. 2023;38:100841.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
30.  Galluzzi L, Aryankalayil MJ, Coleman CN, Formenti SC. Emerging evidence for adapting radiotherapy to immunotherapy. Nat Rev Clin Oncol. 2023;20:543-557.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 164]  [Reference Citation Analysis (0)]
31.  Kim K, Oh D, Noh JM, Min YW, Kim HK, Ahn YC. Outcomes following hypofractionated radiation therapy alone for surgically unfit early esophageal squamous cell carcinoma patients; a retrospective single center analysis. Radiother Oncol. 2024;190:109982.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
32.  Yang L, Murthy S, Cortellini A, Lim EA, Gonzalez M, Pinato DJ, Abdel-Malek M, Mahmoud S, Martin NM. Effects of immune checkpoint inhibitor associated endocrinopathies on cancer survival. Front Endocrinol (Lausanne). 2024;15:1369268.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
33.  Yao L, Jia G, Lu L, Bao Y, Ma W. Factors affecting tumor responders and predictive biomarkers of toxicities in cancer patients treated with immune checkpoint inhibitors. Int Immunopharmacol. 2020;85:106628.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 25]  [Cited by in RCA: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
34.  Chen Q, Guo X, Ma W. Opportunities and challenges of CD47-targeted therapy in cancer immunotherapy. Oncol Res. 2023;32:49-60.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 28]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
35.  Liu Z, Zhang Y, Ma N, Yang Y, Ma Y, Wang F, Wang Y, Wei J, Chen H, Tartarone A, Velotta JB, Dayyani F, Gabriel E, Wakefield CJ, Kidane B, Carbonelli C, Long L, Liu Z, Su J, Li Z. Progenitor-like exhausted SPRY1(+)CD8(+) T cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell. 2023;41:1852-1870.e9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 49]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
36.  Xu S, Wang Q, Ma W. Cytokines and soluble mediators as architects of tumor microenvironment reprogramming in cancer therapy. Cytokine Growth Factor Rev. 2024;76:12-21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
37.  Xu S, Ma Y, Jiang X, Wang Q, Ma W. CD39 transforming cancer therapy by modulating tumor microenvironment. Cancer Lett. 2024;597:217072.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
38.  Liu H, Dong A, Rasteh AM, Wang P, Weng J. Identification of the novel exhausted T cell CD8 + markers in breast cancer. Sci Rep. 2024;14:19142.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 43]  [Cited by in RCA: 59]  [Article Influence: 59.0]  [Reference Citation Analysis (0)]
39.  Tie Y, Tang F, Wei YQ, Wei XW. Immunosuppressive cells in cancer: mechanisms and potential therapeutic targets. J Hematol Oncol. 2022;15:61.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 285]  [Article Influence: 95.0]  [Reference Citation Analysis (0)]
40.  Guo S, Yao Y, Tang Y, Xin Z, Wu D, Ni C, Huang J, Wei Q, Zhang T. Radiation-induced tumor immune microenvironments and potential targets for combination therapy. Signal Transduct Target Ther. 2023;8:205.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 74]  [Reference Citation Analysis (0)]
41.  Zhang Z, Liu X, Chen D, Yu J. Radiotherapy combined with immunotherapy: the dawn of cancer treatment. Signal Transduct Target Ther. 2022;7:258.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 302]  [Reference Citation Analysis (0)]
42.  Lynch C, Pitroda SP, Weichselbaum RR. Radiotherapy, immunity, and immune checkpoint inhibitors. Lancet Oncol. 2024;25:e352-e362.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 35]  [Reference Citation Analysis (0)]
43.  Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys. 2024;119:42-55.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 10]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
44.  Meirovitz A, Gross M, Cohen S, Popovtzer A, Barak V. Effect of irradiation on cytokine production in cancer patients. Int J Biol Markers. 2022;37:360-367.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
45.  Herrera FG, Ronet C, Ochoa de Olza M, Barras D, Crespo I, Andreatta M, Corria-Osorio J, Spill A, Benedetti F, Genolet R, Orcurto A, Imbimbo M, Ghisoni E, Navarro Rodrigo B, Berthold DR, Sarivalasis A, Zaman K, Duran R, Dromain C, Prior J, Schaefer N, Bourhis J, Dimopoulou G, Tsourti Z, Messemaker M, Smith T, Warren SE, Foukas P, Rusakiewicz S, Pittet MJ, Zimmermann S, Sempoux C, Dafni U, Harari A, Kandalaft LE, Carmona SJ, Dangaj Laniti D, Irving M, Coukos G. Low-Dose Radiotherapy Reverses Tumor Immune Desertification and Resistance to Immunotherapy. Cancer Discov. 2022;12:108-133.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 54]  [Cited by in RCA: 264]  [Article Influence: 88.0]  [Reference Citation Analysis (0)]
46.  Chen N, Xu X, Fan Y. Immune checkpoint inhibitors in the treatment of oesophageal squamous cell carcinoma: where are we and where are we going? Ther Adv Med Oncol. 2023;15:17588359231189420.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
47.  Naing A, Hajjar J, Gulley JL, Atkins MB, Ciliberto G, Meric-Bernstam F, Hwu P. Strategies for improving the management of immune-related adverse events. J Immunother Cancer. 2020;8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 58]  [Cited by in RCA: 59]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
48.  Schneider BJ, Naidoo J, Santomasso BD, Lacchetti C, Adkins S, Anadkat M, Atkins MB, Brassil KJ, Caterino JM, Chau I, Davies MJ, Ernstoff MS, Fecher L, Ghosh M, Jaiyesimi I, Mammen JS, Naing A, Nastoupil LJ, Phillips T, Porter LD, Reichner CA, Seigel C, Song JM, Spira A, Suarez-Almazor M, Swami U, Thompson JA, Vikas P, Wang Y, Weber JS, Funchain P, Bollin K. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J Clin Oncol. 2021;39:4073-4126.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 115]  [Cited by in RCA: 987]  [Article Influence: 246.8]  [Reference Citation Analysis (0)]
49.  Hu LL, Rong F, Liu L, Zhang L, Zhang LL, Yang Q, Xia ZL, Wang H. Prognosis of radiotherapy for esophageal cancer in elderly patients exceeding seventy-five years old. World J Gastrointest Oncol. 2024;16:4636-4649.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
50.  Tosca EM, De Carlo A, Ronchi D, Magni P. Model-Informed Reinforcement Learning for Enabling Precision Dosing Via Adaptive Dosing. Clin Pharmacol Ther. 2024;116:619-636.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
51.  Zhang X, Cai X, Yan C. Opportunities and challenges in combining immunotherapy and radiotherapy in esophageal cancer. J Cancer Res Clin Oncol. 2023;149:18253-18270.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
52.  Jeon SH, Song C, Eom KY, Kim IA, Kim JS. Modulation of CD8(+) T Cell Responses by Radiotherapy-Current Evidence and Rationale for Combination with Immune Checkpoint Inhibitors. Int J Mol Sci. 2023;24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
53.  Huang S, Xu J, Baran N, Ma W. Advancing the next generation of cancer treatment with circular RNAs in CAR-T cell therapy. Biomed Pharmacother. 2024;181:117753.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
54.  Cianciotti BC, Magnani ZI, Ugolini A, Camisa B, Merelli I, Vavassori V, Potenza A, Imparato A, Manfredi F, Abbati D, Perani L, Spinelli A, Shifrut E, Ciceri F, Vago L, Di Micco R, Naldini L, Genovese P, Ruggiero E, Bonini C. TIM-3, LAG-3, or 2B4 gene disruptions increase the anti-tumor response of engineered T cells. Front Immunol. 2024;15:1315283.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
55.  Yang MQ, Zhang SL, Sun L, Huang LT, Yu J, Zhang JH, Tian Y, Han CB, Ma JT. Targeting mitochondria: restoring the antitumor efficacy of exhausted T cells. Mol Cancer. 2024;23:260.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
56.  Qiu X, Yu Z, Lu X, Jin X, Zhu J, Zhang R. PD-1 and LAG-3 dual blockade: emerging mechanisms and potential therapeutic prospects in cancer. Cancer Biol Med. 2024;21:970-976.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
57.  Zabeti Touchaei A, Vahidi S. MicroRNAs as regulators of immune checkpoints in cancer immunotherapy: targeting PD-1/PD-L1 and CTLA-4 pathways. Cancer Cell Int. 2024;24:102.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 37]  [Reference Citation Analysis (0)]
58.  Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther. 2024;9:176.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 62]  [Cited by in RCA: 81]  [Article Influence: 81.0]  [Reference Citation Analysis (0)]
59.  Rudqvist NP, Charpentier M, Lhuillier C, Wennerberg E, Spada S, Sheridan C, Zhou XK, Zhang T, Formenti SC, Sims JS, Alonso A, Demaria S. Immunotherapy targeting different immune compartments in combination with radiation therapy induces regression of resistant tumors. Nat Commun. 2023;14:5146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 30]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
60.  Pan X, Zhang W, Guo H, Wang L, Wu H, Ding L, Yang B. Strategies involving STING pathway activation for cancer immunotherapy: Mechanism and agonists. Biochem Pharmacol. 2023;213:115596.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
61.  Berland L, Gabr Z, Chang M, Ilié M, Hofman V, Rignol G, Ghiringhelli F, Mograbi B, Rashidian M, Hofman P. Further knowledge and developments in resistance mechanisms to immune checkpoint inhibitors. Front Immunol. 2024;15:1384121.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
62.  Ji X, Jiang W, Wang J, Zhou B, Ding W, Liu S, Huang H, Chen G, Sun X. Application of individualized multimodal radiotherapy combined with immunotherapy in metastatic tumors. Front Immunol. 2022;13:1106644.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
63.  Sugawara K, Fukuda T, Murakami C, Oka D, Yoshii T, Amori G, Ishibashi K, Kobayashi Y, Hara H, Kanda H, Motoi N. Impacts of tumor microenvironment during neoadjuvant chemotherapy in patients with esophageal squamous cell carcinoma. Cancer Sci. 2024;115:2819-2830.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (1)]
64.  Liu L, Liu Y, Xu L, Ding Y, Han J, Wang Q, Chen X, Almhanna K, Han C, Wang L. Tumor immune microenvironment remodeling and prognosis of patients with esophageal squamous cell carcinoma after neoadjuvant chemotherapy with and without immunotherapy: a retrospective cohort study. J Thorac Dis. 2024;16:3909-3922.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
65.  Chen B, Liu S, Zhu Y, Wang R, Cheng X, Chen B, Dragomir MP, Zhang Y, Hu Y, Liu M, Li Q, Yang H, Xi M. Predictive role of ctDNA in esophageal squamous cell carcinoma receiving definitive chemoradiotherapy combined with toripalimab. Nat Commun. 2024;15:1919.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
66.  Yu B, Ma W. Biomarker discovery in hepatocellular carcinoma (HCC) for personalized treatment and enhanced prognosis. Cytokine Growth Factor Rev. 2024;79:29-38.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 14]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
67.  Noori M, Yousefi AM, Zali MR, Bashash D. Predictive value of PD-L1 expression in response to immune checkpoint inhibitors for esophageal cancer treatment: A systematic review and meta-analysis. Front Oncol. 2022;12:1021859.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
68.  Bao Y, Zhang D, Guo H, Ma W. Beyond blood: Advancing the frontiers of liquid biopsy in oncology and personalized medicine. Cancer Sci. 2024;115:1060-1072.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 23]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
69.  Al-Showbaki L, Wilson B, Tamimi F, Molto C, Mittal A, Cescon DW, Amir E. Changes in circulating tumor DNA and outcomes in solid tumors treated with immune checkpoint inhibitors: a systematic review. J Immunother Cancer. 2023;11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
70.  Yu B, Shao S, Ma W. Frontiers in pancreatic cancer on biomarkers, microenvironment, and immunotherapy. Cancer Lett. 2025;610:217350.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 14]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
71.  Han J, Guo X, Zhao L, Zhang H, Ma S, Li Y, Zhao D, Wang J, Xue F. Development and Validation of Esophageal Squamous Cell Carcinoma Risk Prediction Models Based on an Endoscopic Screening Program. JAMA Netw Open. 2023;6:e2253148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
72.  Shang X, Xie Y, Yu J, Zhang C, Zhao G, Liang F, Liu L, Zhang W, Li R, Yu W, Yue J, Chen C, Duan X, Ma Z, Chen Z, Xiong Y, Yang F, Xiao J, Zhang R, Liu P, Cheng Y, Cao F, Guo F, Liu G, Meng B, Zhou D, Sun Y, Ren X, Yu J, Hao J, Jiang H. A prospective study of neoadjuvant pembrolizumab plus chemotherapy for resectable esophageal squamous cell carcinoma: The Keystone-001 trial. Cancer Cell. 2024;42:1747-1763.e7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
73.  Kim S. Overview of clinical study designs. Clin Exp Emerg Med. 2024;11:33-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
74.  Andrade C. Sample Size and its Importance in Research. Indian J Psychol Med. 2020;42:102-103.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 137]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
75.  Serdar CC, Cihan M, Yücel D, Serdar MA. Sample size, power and effect size revisited: simplified and practical approaches in pre-clinical, clinical and laboratory studies. Biochem Med (Zagreb). 2021;31:010502.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 386]  [Cited by in RCA: 608]  [Article Influence: 152.0]  [Reference Citation Analysis (0)]
76.  Buse JB, Austin CP, Johnston SC, Lewis-Hall F, March AN, Shore CK, Tenaerts P, Rutter JL. A framework for assessing clinical trial site readiness. J Clin Transl Sci. 2023;7:e151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
77.  Han R, Acosta JN, Shakeri Z, Ioannidis JPA, Topol EJ, Rajpurkar P. Randomised controlled trials evaluating artificial intelligence in clinical practice: a scoping review. Lancet Digit Health. 2024;6:e367-e373.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 81]  [Cited by in RCA: 68]  [Article Influence: 68.0]  [Reference Citation Analysis (0)]
78.  Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther. 2024;9:132.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 151]  [Reference Citation Analysis (0)]
79.  Yin X, Song Y, Deng W, Blake N, Luo X, Meng J. Potential predictive biomarkers in antitumor immunotherapy: navigating the future of antitumor treatment and immune checkpoint inhibitor efficacy. Front Oncol. 2024;14:1483454.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
80.  Sachdeva M, Zhao JJ, Zhu KX, Yap DTW, Wong NZH, Kumarakulasinghe NB, Tey J, Sundar R. PD-L1 Expression as a Negative Predictive Biomarker in Advanced Esophageal Squamous-Cell Cancer Treated with Chemotherapy Alone. ESMO Gastrointest Oncol. 2025;7.  [PubMed]  [DOI]  [Full Text]
81.  Zhao X, Bao Y, Meng B, Xu Z, Li S, Wang X, Hou R, Ma W, Liu D, Zheng J, Shi M. From rough to precise: PD-L1 evaluation for predicting the efficacy of PD-1/PD-L1 blockades. Front Immunol. 2022;13:920021.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 35]  [Reference Citation Analysis (0)]
82.  Ng HY, Ko JMY, Lam KO, Kwong DLW, Lo AWI, Wong IYH, Wong CLY, Chan SY, Chan KK, Law TT, Dai W, Fong HCH, Choy FSF, Lo CK, Chen C, Law SYK, Lung ML. Circulating Tumor DNA Dynamics as Prognostic Markers in Locally Advanced and Metastatic Esophageal Squamous Cell Carcinoma. JAMA Surg. 2023;158:1141-1150.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 16]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
83.  Vellanki PJ, Ghosh S, Pathak A, Fusco MJ, Bloomquist EW, Tang S, Singh H, Philip R, Pazdur R, Beaver JA. Regulatory implications of ctDNA in immuno-oncology for solid tumors. J Immunother Cancer. 2023;11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 32]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
84.  Bakaloudi DR, Talukder R, Makrakis D, Diamantopoulos L, Enright T, Leary JB, Patgunarajah U, Thomas VM, Swami U, Agarwal N, Jindal T, Koshkin VS, Brown JR, Barata P, Murgić J, Miletić M, Johnson J, Zakharia Y, Hui G, Drakaki A, Duran I, Buznego LA, Barrera RM, Castañeda DM, Rey-Cárdenas M, Castellano D, Nguyen CB, Park JJ, Alva A, McKay RR, Stewart TF, Epstein IB, Bellmunt J, Wright JL, Gupta S, Grivas P, Khaki AR. Association of Tumor Mutational Burden and Microsatellite Instability With Response and Outcomes in Patients With Urothelial Carcinoma Treated With Immune Checkpoint Inhibitor. Clin Genitourin Cancer. 2024;22:102198.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Reference Citation Analysis (0)]
85.  Ju C, He J, Wang C, Sheng J, Jia J, Du D, Li H, Zhou M, He F. Current advances and future perspectives on the functional roles and clinical implications of circular RNAs in esophageal squamous cell carcinoma: more influential than expected. Biomark Res. 2022;10:41.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
86.  Reese KL, Pantel K, Smit DJ. Multibiomarker panels in liquid biopsy for early detection of pancreatic cancer - a comprehensive review. J Exp Clin Cancer Res. 2024;43:250.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
87.  Shao S, Risch E, Burner D, Lu L, Minev B, Ma W. IFNγ enhances cytotoxic efficiency of the cytotoxic T lymphocytes against human glioma cells. Int Immunopharmacol. 2017;47:159-165.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 20]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
88.  Yang W, Liu S, Mao M, Gong Y, Li X, Lei T, Liu C, Wu S, Hu Q. T-cell infiltration and its regulatory mechanisms in cancers: insights at single-cell resolution. J Exp Clin Cancer Res. 2024;43:38.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 28]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
89.  Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther. 2024;9:274.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 75]  [Reference Citation Analysis (0)]
90.  Colomer R, Miranda J, Romero-Laorden N, Hornedo J, González-Cortijo L, Mouron S, Bueno MJ, Mondéjar R, Quintela-Fandino M. Usefulness and real-world outcomes of next generation sequencing testing in patients with cancer: an observational study on the impact of selection based on clinical judgement. EClinicalMedicine. 2023;60:102029.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 32]  [Reference Citation Analysis (0)]
91.  Jardim DL, Goodman A, de Melo Gagliato D, Kurzrock R. The Challenges of Tumor Mutational Burden as an Immunotherapy Biomarker. Cancer Cell. 2021;39:154-173.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 349]  [Cited by in RCA: 731]  [Article Influence: 182.8]  [Reference Citation Analysis (0)]
92.  Lv J, Xu LX, Li ZX, Lin L, Wu CF, Quan TQ, Zhen ZC, Li WF, Tang LL, Mao YP, Chen L, Guo R, Zhang LL, Ai XL, Wu SY, Hao MY, Wei D, Li JB, Ma J, Chen YP, Zhou GQ, Sun Y. Longitudinal on-treatment circulating tumor DNA as a biomarker for real-time dynamic risk monitoring in cancer patients: The EP-SEASON study. Cancer Cell. 2024;42:1401-1414.e4.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 17]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
93.  Qin Y, Huo M, Liu X, Li SC. Biomarkers and computational models for predicting efficacy to tumor ICI immunotherapy. Front Immunol. 2024;15:1368749.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 13]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
94.  Gao W, Liu S, Wu Y, Wei W, Yang Q, Li W, Chen H, Luo A, Wang Y, Liu Z. Enhancer demethylation-regulated gene score identified molecular subtypes, inspiring immunotherapy or CDK4/6 inhibitor therapy in oesophageal squamous cell carcinoma. EBioMedicine. 2024;105:105177.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
95.  Benbow JH, Rivera DR, Lund JL, Feldman JE, Kim ES. Increasing Inclusiveness of Patient-Centric Clinical Evidence Generation in Oncology: Real-World Data and Clinical Trials. Am Soc Clin Oncol Educ Book. 2022;42:1-11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
96.  Puhr HC, Prager GW, Ilhan-Mutlu A. How we treat esophageal squamous cell carcinoma. ESMO Open. 2023;8:100789.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 57]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
97.  Kelly RJ. Immunotherapy for Esophageal and Gastric Cancer. Am Soc Clin Oncol Educ Book. 2017;37:292-300.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 53]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
98.  Liu C, Sun H, Huang W, Wang Z, Fu C, Han D, Zhao Q, Wu X, Li B. Sintilimab as maintenance treatment for local/regional recurrent esophageal squamous carcinoma after concurrent chemoradiotherapy: a single-arm Ib/II phase study. Front Immunol. 2023;14:1193394.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
99.  Sasaki M, Tamamura H, Tameshige Y, Azuma Y, Maeda Y, Matsushita K, Sato Y, Takamatsu S, Inoue K, Tabata Y, Yoshimura H, Yamamoto K. Dose Evaluation in 2-Phase Method for Advanced Esophageal Cancer by Hybrid Irradiation Techniques. Int J Part Ther. 2024;11:100010.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
100.  Dona Lemus OM, Cao M, Cai B, Cummings M, Zheng D. Adaptive Radiotherapy: Next-Generation Radiotherapy. Cancers (Basel). 2024;16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 33]  [Cited by in RCA: 26]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
101.  Powers C, Kaya E, Bertinetti A, Hung A. The current state of proton radiotherapy. Curr Probl Cancer. 2024;53:101153.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
102.  Yu P, Zhu C, You X, Gu W, Wang X, Wang Y, Bu R, Wang K. The combination of immune checkpoint inhibitors and antibody-drug conjugates in the treatment of urogenital tumors: a review insights from phase 2 and 3 studies. Cell Death Dis. 2024;15:433.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 13]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
103.  Garg P, Pareek S, Kulkarni P, Horne D, Salgia R, Singhal SS. Next-Generation Immunotherapy: Advancing Clinical Applications in Cancer Treatment. J Clin Med. 2024;13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
104.  Hoefnagel SJM, Boonstra JJ, Russchen MJAM, Krishnadath KK. Towards Personalized Treatment Strategies for Esophageal Adenocarcinoma; A Review on the Molecular Characterization of Esophageal Adenocarcinoma and Current Research Efforts on Individualized Curative Treatment Regimens. Cancers (Basel). 2021;13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
105.  Liu B, Zhou H, Tan L, Siu KTH, Guan XY. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther. 2024;9:175.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 182]  [Article Influence: 182.0]  [Reference Citation Analysis (0)]
106.  Liu S, Wang W, Hu S, Jia B, Tuo B, Sun H, Wang Q, Liu Y, Sun Z. Radiotherapy remodels the tumor microenvironment for enhancing immunotherapeutic sensitivity. Cell Death Dis. 2023;14:679.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 42]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
107.  Hu A, Sun L, Lin H, Liao Y, Yang H, Mao Y. Harnessing innate immune pathways for therapeutic advancement in cancer. Signal Transduct Target Ther. 2024;9:68.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 65]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
108.  Liu Y, Wu W, Cai C, Zhang H, Shen H, Han Y. Patient-derived xenograft models in cancer therapy: technologies and applications. Signal Transduct Target Ther. 2023;8:160.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 163]  [Cited by in RCA: 163]  [Article Influence: 81.5]  [Reference Citation Analysis (0)]
109.  Catanzaro E, Beltrán-Visiedo M, Galluzzi L, Krysko DV. Immunogenicity of cell death and cancer immunotherapy with immune checkpoint inhibitors. Cell Mol Immunol. 2025;22:24-39.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 16]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
110.  Wu P, Qin G, Liu J, Zhao Q, Zhao X, Song X, Wang L, Yang S, Zhang Y. Distinct immune signatures for predicting the immunotherapy efficacy of esophageal squamous cell carcinoma or adenocarcinoma. Cancer Immunol Immunother. 2025;74:47.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
111.  Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther. 2024;9:308.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 31]  [Reference Citation Analysis (0)]
112.  Liu H, Wang P. CRISPR screening and cell line IC50 data reveal novel key genes for trametinib resistance. Clin Exp Med. 2024;25:21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 15]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
113.  Wang Z, Xiao Z, Zhang T, Lu M, Li H, Cao J, Zheng J, Zhou Y, Dai J, Wang C, Chen L, Xu J. Development and validation of a novel artificial intelligence algorithm for precise prediction the postoperative prognosis of esophageal squamous cell carcinoma. BMC Cancer. 2025;25:134.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
114.  Passaro A, Al Bakir M, Hamilton EG, Diehn M, André F, Roy-Chowdhuri S, Mountzios G, Wistuba II, Swanton C, Peters S. Cancer biomarkers: Emerging trends and clinical implications for personalized treatment. Cell. 2024;187:1617-1635.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 144]  [Article Influence: 144.0]  [Reference Citation Analysis (0)]
115.  Shea L, Pesa J, Geonnotti G, Powell V, Kahn C, Peters W. Improving diversity in study participation: Patient perspectives on barriers, racial differences and the role of communities. Health Expect. 2022;25:1979-1987.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 46]  [Reference Citation Analysis (0)]
116.  Gross AS, Harry AC, Clifton CS, Della Pasqua O. Clinical trial diversity: An opportunity for improved insight into the determinants of variability in drug response. Br J Clin Pharmacol. 2022;88:2700-2717.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 32]  [Reference Citation Analysis (0)]
117.  Tomiwa T, Wong E, Miller HN, Ogungbe O, Byiringiro S, Plante T, Himmelfarb CR. Leveraging digital tools to enhance diversity and inclusion in clinical trial recruitment. Front Public Health. 2024;12:1483367.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
118.  Khalifa M, Albadawy M. Artificial Intelligence for Clinical Prediction: Exploring Key Domains and Essential Functions. Comput Methods Programs Biomed Update. 2024;5:100148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 27]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
119.  Cuzick J. The importance of long-term follow up of participants in clinical trials. Br J Cancer. 2023;128:432-438.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 29]  [Reference Citation Analysis (0)]
120.  Oshima K, Tsushima T, Ito Y, Kato K. Recent progress in chemoradiotherapy for oesophageal squamous cell carcinoma. Jpn J Clin Oncol. 2024;54:395-402.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
121.  Mahumud RA, Shahjalal M, Dahal PK, Mosharaf MP, Hoque ME, Wawryk O. Systemic therapy and radiotherapy related complications and subsequent hospitalisation rates: a systematic review. BMC Cancer. 2024;24:826.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
122.  Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther. 2024;9:57.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 21]  [Cited by in RCA: 57]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
123.  Finney Rutten LJ, Ridgeway JL, Griffin JM. Advancing Translation of Clinical Research Into Practice and Population Health Impact Through Implementation Science. Mayo Clin Proc. 2024;99:665-676.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
124.  Zhang Z, Leng Z, Fang K, Sun M, Li Z, Kang L, Chen T, Xu M. Global research trend of esophageal squamous cell carcinoma from 2012 to 2022: a bibliometric analysis. Front Oncol. 2022;12:977935.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]