Skip to main content

MINI REVIEW article

Front. Immunol., 30 October 2024
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Harnessing Single-Cell Insights: Pioneering Predictive Markers for Immunotherapy Efficacy in Solid Tumors View all 8 articles

A single-cell perspective on immunotherapy for pancreatic cancer: from microenvironment analysis to therapeutic strategy innovation

Rui Wang,,&#x;Rui Wang1,2,3†Jie Liu,&#x;Jie Liu1,2†Bo Jiang,&#x;Bo Jiang1,2†Benjian Gao,&#x;Benjian Gao1,2†Honghao LuoHonghao Luo4Fengyi Yang,Fengyi Yang1,2Yuntao Ye,Yuntao Ye1,2Zhuo Chen,Zhuo Chen1,2Hong Liu,Hong Liu1,2Cheng Cui,Cheng Cui1,2Ke Xu*Ke Xu5*Bo Li,*Bo Li1,2*Xiaoli Yang,*Xiaoli Yang1,2*
  • 1Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
  • 2Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, China
  • 3General Surgery Day Ward, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
  • 4Department of Radiology, Xichong People’s Hospital, Nanchong, China
  • 5Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China

Pancreatic cancer remains one of the most lethal malignancies, with conventional treatment options providing limited efficacy. Recent advancements in immunotherapy have offered new hope, yet the unique tumor microenvironment (TME) of pancreatic cancer poses significant challenges to its successful application. This review explores the transformative impact of single-cell technology on the understanding and treatment of pancreatic cancer. By enabling high-resolution analysis of cellular heterogeneity within the TME, single-cell approaches have elucidated the complex interplay between various immune and tumor cell populations. These insights have led to the identification of predictive biomarkers and the development of innovative, personalized immunotherapeutic strategies. The review discusses the role of single-cell technology in dissecting the intricate immune landscape of pancreatic cancer, highlighting the discovery of T cell exhaustion profiles and macrophage polarization states that influence treatment response. Moreover, it outlines the potential of single-cell data in guiding the selection of immunotherapy drugs and optimizing treatment plans. The review also addresses the challenges and prospects of translating these single-cell-based innovations into clinical practice, emphasizing the need for interdisciplinary research and the integration of artificial intelligence to overcome current limitations. Ultimately, the review underscores the promise of single-cell technology in driving therapeutic strategy innovation and improving patient outcomes in the battle against pancreatic cancer.

1 Background

Pancreatic cancer has emerged as a focal point in global medical research due to its exceedingly high fatality rate and the scarcity of treatment options (14). This type of cancer’s five-year survival rate is astonishingly low, less than 10%, significantly lower than most other types of cancer (5). These poor survival rates have not changed significantly in nearly 40 years, highlighting the malignancy of pancreatic cancer and the urgency of treatment. One of the primary challenges of pancreatic cancer lies in its almost asymptomatic early stages, leading to a diagnosis at an advanced stage for the majority of patients (6, 7). Furthermore, the difficulty in diagnosing pancreatic cancer and the lack of early diagnosis mean that patients miss the optimal time to receive effective treatment (8, 9). About 70% of pancreatic cancer patients will develop cachexia that cannot be reversed through traditional nutritional support (10). Consequently, researchers aim to improve early diagnosis rates and find more effective treatments to enhance survival and quality of life.

Immunotherapy has shown significant potential in cancer treatment, leveraging the patient’s immune system to recognize and eliminate cancer cells (11). Unlike traditional surgery, chemotherapy, and radiation therapy, immunotherapy activates the immune system to target cancer cells effectively. Although successful in treating cancers like melanoma and non-small cell lung cancer, immunotherapy has lagged in pancreatic cancer due to unique tumor microenvironment and immune evasion mechanisms (12, 13).Pancreatic cancer’s dense desmoplastic stroma hinders immune cell infiltration, a stark contrast to the less dense environments in cancers like breast or prostate cancer (14). Additionally, pancreatic cancers secrete high levels of immunosuppressive cytokines such as TGF-β and IL-10, creating a tumor microenvironment that hinders effective T-cell response (15, 16). The lower mutational burden in pancreatic cancer results in fewer targets for immune recognition compared to other cancers with high mutational burdens (17). Moreover, pancreatic cancer cells express high levels of PD-L1, leading to T-cell anergy and apoptosis (18, 19). However, with in-depth research into the pancreatic cancer tumor microenvironment, scientists have begun to find that adjusting the use of immune checkpoint inhibitors can effectively change the tumor microenvironment, enhancing the immune system’s ability to attack pancreatic cancer cells (20). Furthermore, combining immunotherapy with other treatments, such as targeted therapy and chemotherapy, has also shown potential to improve treatment effects, providing new hope for pancreatic cancer patients and opening new directions for future treatment research (2123).

Single-cell technology provides a novel perspective for understanding the complexity of pancreatic cancer. This technology enables high-throughput gene expression analysis at the single-cell level, revealing the cellular heterogeneity in the tumor microenvironment (24, 25). Cellular-level heterogeneity is a significant challenge in treating pancreatic cancer, as it leads to varied responses to treatments (26). Single-cell technology has identified distinct subpopulations of cancer-associated fibroblasts that contribute differently to tumor progression and therapy resistance. For instance, Elyada et al. identified fibroblast subtypes with pro-tumorigenic and anti-tumorigenic properties, which has significant implications for targeted therapies. Peng et al. used scRNA-seq(single-cell RNA sequencing) to uncover rare cell populations resistant to conventional therapies, providing new therapeutic targets (27).

Through single-cell technology, researchers can analyze gene expression patterns, functional states, and interactions of different cell types in the tumor microenvironment, crucial for understanding pancreatic cancer’s response to immunotherapy (28). Specifically, this technology helps identify tumor cells evading immune surveillance and immune cells playing key roles in tumor combat, providing a basis for designing new immunotherapy strategies (29). For example, Zhang et al. used single-cell analysis to identify immune cell subsets correlating with better patient outcomes, paving the way for personalized immunotherapy (30). Additionally, single-cell technology can monitor treatment effects by analyzing changes in tumor microenvironment cells before and after treatment, assessing treatment plans’ effectiveness (20, 3134).

2 The revolutionary impact of single-cell technology

2.1 Application of single-cell technology in pancreatic cancer research

Single-cell technology allows detailed study of each cancer cell, revealing unknown molecular markers and pathways. For example, through single-cell sequencing technology, researchers have been able to depict the signaling pathway activity map in pancreatic cancer cells, revealing differences between different cell populations (35). These differences are not only present among cancer cells expressing KRAS mutations but also among cells in the tumor microenvironment, such as macrophages and T cells, in terms of their functional states (36). Through these in-depth analyses, researchers have discovered that cells in the tumor microenvironment interact with cancer cells by secreting various factors (such as cytokines and chemokines), which are crucial for tumor growth, metastasis, and response to treatment (37, 38).

2.2 Single-cell analysis reveals tumor heterogeneity and immune microenvironment

Single-cell technology reveals not only tumor cell heterogeneity but also the complexity of immune cells in the tumor microenvironment (39). These cells play dual roles in tumor growth and spread. Single-cell sequencing helps identify and classify immune cell subtypes in the tumor and study their interactions with cancer cells through specific molecular pathways. For example, Tregs (regulatory T cells) suppress immune response by expressing molecules like CTLA-4 and TGF-β, facilitating tumor cell escape (40). This insight supports developing therapeutic strategies targeting these immunosuppressive pathways. Clinical trials are exploring inhibitors of CTLA-4 and TGF-β for pancreatic cancer, with early-phase studies indicating that combining these inhibitors with immune checkpoint blockade can enhance anti-tumor immune responses and improve outcomes (41). Similarly, analysis of macrophages in the tumor microenvironment shows that M2-type macrophages promote tumor growth and suppress immune responses by secreting molecules like IL-10 and upregulating PD-L1, aiding tumor cells in evading immune surveillance (42, 43). Therapeutic strategies are being developed to reprogram these M2 macrophages into the pro-inflammatory M1 phenotype, supporting anti-tumor immunity. Preclinical studies show that targeting IL-10 and PD-L1, combined with macrophage reprogramming agents, can reduce tumor burden and enhance treatment efficacy (19). These examples illustrate the crucial role of single-cell technology in revealing the complexity of pancreatic cancer and its microenvironment, providing unprecedented opportunities to understand disease mechanisms and develop new therapeutic methods.

3 Single-cell analysis of the pancreatic cancer immune microenvironment

3.1 Complexity of the pancreatic cancer immune microenvironment and its impact on treatment response

The pancreatic cancer immune microenvironment is a complex network of tumor cells, immune cells, fibroblasts, and endothelial cells. Their interactions affect tumor development and treatment response. Tregs and TAMs (tumor-associated macrophages), prevalent in the pancreatic cancer immune microenvironment, suppress effector T cell activity and promote immune evasion by releasing factors like TGF-β (44, 45). Particularly, TAMs in pancreatic cancer often exhibit an M2 polarization state, where M2-type TAMs further promote tumor growth and angiogenesis by secreting molecules like IL-10 and VEGF (4648). Moreover, the fibrotic characteristic of pancreatic cancer, namely the formation of scar tissue and dense deposition of the extracellular matrix, constitutes a physical and biochemical barrier that hinders the infiltration of immune cells. This feature not only reduces the permeability of tumor tissue to drugs but also limits the access and attack capabilities of immune cells to tumor cells (4951) (Figure 1).

Figure 1
www.frontiersin.org

Figure 1. Single-cell perspective of immune cell populations and tumor cell populations in the pancreatic cancer microenvironment.

3.2 How single-cell data can help identify mechanisms of immune therapy response

Single-cell technology, particularly scRNA-seq, provides a powerful tool for deciphering the pancreatic cancer immune microenvironment, enabling researchers to study cellular heterogeneity and complex interactions at an unprecedented resolution (52, 53). Through single-cell analysis of pancreatic cancer patient samples, researchers have identified a group of CD8+ T cells specifically expressing PD-1, which are exceedingly rare in untreated tumors but significantly increase after PD-1/PD-L1 blockade therapy (54, 55). This finding not only confirms the role of PD-1 as an immune checkpoint but also identifies specific cell subpopulations responding to immunotherapy, offering potential for personalized medicine. Further, single-cell sequencing reveals “immune desert” phenomena where some pancreatic tumors have scarce immune cells, actively repelled by specific chemokine and adhesion molecule patterns (56, 57). This rich analysis helps understand the complexity of the pancreatic cancer immune microenvironment and reveals mechanisms of immune therapy response and failure, crucial for designing new therapeutic strategies and achieving personalized treatment (58).

4 Predictive biomarkers discovered through single-cell technology

4.1 Predictive biomarkers of immune therapy response identified by single-cell analysis

Single-cell technology has revealed key biomarkers predicting immune therapy response in pancreatic cancer. High-resolution scRNA-seq allows precise analysis of cellular heterogeneity in the tumor immune microenvironment, identifying biomarkers like high PD-1 expression on CD8+ T cells, indicating an exhausted state linked to responsiveness to anti-PD-1/PD-L1 therapy (56, 5860). Clinically, these findings can help personalize therapy, prioritizing patients with high PD-1 levels for anti-PD-1/PD-L1 treatment (61). The M2 polarization state of TAMs, secreting IL-10 and TGF-β, also impacts immune therapy effectiveness (62). Identifying these states through single-cell sequencing can guide combination therapies targeting PD-1/PD-L1 and modulating the macrophage environment to enhance immune response (63). Single-cell data also reveal signaling pathways regulating immune cell activity, providing mechanisms to predict immune therapy outcomes accurately (6466).

4.2 Exploring the potential contributions of these markers to personalized treatment strategies

The predictive markers discovered by single-cell technology provide a substantial scientific basis for personalizing pancreatic cancer treatment. In clinical practice, the presence of these markers can guide physicians in selecting the most suitable immune therapy drugs, optimizing treatment plans, and avoiding the waste of resources on treatments unlikely to be effective. For example, by pre-testing tumor samples for specific phenotypes and functional states of immune cells, doctors can predict the patient’s responsiveness to PD-1/PD-L1 or CTLA-4 inhibitors, thus selectively choosing the treatment method most likely to benefit (67). Additionally, these in-depth insights obtained through single-cell analysis also offer directions for developing new therapeutic targets and strategies (25, 42). For instance, targeting specific subsets of exhausted T cells found in the pancreatic cancer tumor microenvironment, researchers can design novel treatment approaches, such as using biologics to restore these cells’ activity or enhance their tumor-killing capabilities (68). Similarly, therapeutic strategies targeting M2-type TAMs could involve transforming these cells into an anti-tumor M1 polarization state to inhibit tumor growth (29, 69, 70).

5 Therapeutic strategy innovation driven by single-cell technology

5.1 New immunotherapy strategies developed based on single-cell analysis results

Single-cell technology, particularly scRNA-seq, has driven innovation and precision in pancreatic cancer treatment strategies (25, 53). By analyzing individual cells in the tumor microenvironment, researchers have identified various cell markers and molecular pathways that predict treatment response. These include specific T cell subtypes, immune checkpoint expression patterns, and levels of pro-inflammatory and anti-inflammatory cytokines (54, 71, 72). Based on these findings, new immunotherapy strategies have emerged, including biomarker-guided immune checkpoint inhibitor therapy, targeted cell therapy, and methods to modulate the tumor microenvironment to enhance immune response (73). For example, Chen et al. (2023) found that integrating immune checkpoint inhibitors with therapies targeting the KRAS pathway significantly enhanced immune response in pancreatic cancer models, reducing tumor progression and extending survival compared to single-agent treatments (74). Additionally, single-cell analysis has revealed aberrant signaling pathways that offer new combination therapy opportunities. Combining specific pathway inhibitors with immune therapies, such as PD-1/PD-L1 blockers, can overcome pancreatic cancer’s immune evasion mechanisms, improving treatment outcomes (75, 76). For instance, Lu et al. found that targeting the JAK/STAT signaling pathway, in conjunction with PD-L1 blockade, enhanced CD8+ T cell infiltration and activity in pancreatic tumors, significantly reducing tumor burden (77).

5.2 Clinical prospects of single-cell-driven therapeutic strategy innovations

Translating single-cell-driven pancreatic cancer immunotherapy strategies from the laboratory to clinical application presents enormous prospects (42). Personalized treatment plans, based on detailed tumor microenvironment analysis, can improve treatment specificity and effectiveness, reduce unnecessary side effects, and enhance patient prognosis and quality of life (67). However, the clinical implementation of these strategies faces numerous challenges. First, the high costs and technical requirements limit the widespread clinical application of single-cell technology (78). Second, although single-cell analysis can provide detailed information about the tumor microenvironment, translating these complex data into effective treatment plans remains a challenge (79). Furthermore, the safety and effectiveness of these single-cell analysis-based treatment strategies need to be verified through clinical trials, a time and resource-intensive process (42). Lastly, the high heterogeneity and complexity of pancreatic cancer mean that even the most advanced treatment strategies may encounter resistance issues, necessitating continuous exploration and optimization of treatment plans (80).

6 Future research directions and challenges

6.1 Deepening exploration of future directions in pancreatic cancer research with single-cell technology

As single-cell technology continues to advance, the exploration of new markers will remain a focus of future research, especially those that can guide immunotherapy and precision medicine, such as specific subtypes of immune cells or unique molecular patterns of tumor cells (25). Additionally, future pancreatic cancer research may emphasize analyzing cell heterogeneity and interactions to reveal subtle differences in cancer development and treatment response (42). Specifically, the combined use of single-cell transcriptomics, single-cell epigenetics, and single-cell proteomics is expected to provide new perspectives on complex molecular mechanisms in pancreatic cancer that are not yet fully understood (25). For example, by analyzing the epigenetic states of different cell types in pancreatic cancer tumors, researchers can identify key epigenetic modifications that promote tumor growth and immune evasion, exploring targeted treatment strategies against these modifications (53). Additionally, Single-cell analysis of cell-cell communication, such as exosome-mediated signaling, will reveal immune suppression network mechanisms, offering strategies to disrupt these networks (81). The application of single-cell technology will not be limited to studying pancreatic cancer cells themselves but will also extend to researching non-tumor cells in the tumor microenvironment, such as immune cells, fibroblasts, and endothelial cells (80, 82). For instance, detailed analysis of immune cell diversity and functional states will uncover immune surveillance evasion mechanisms, discovering new immune checkpoints or immunoregulatory molecules as therapeutic targets (52, 54).

6.2 Challenges and acceleration of pancreatic cancer treatment progress through interdisciplinary research

Interdisciplinary research, including AI and gene editing technology, plays a crucial role in advancing single-cell studies. AI, especially deep learning, has great potential in processing and analyzing large-scale biological data (83, 84). AI can predict treatment responses at the cellular level, helping design suitable treatment plans for individual disease characteristics. For instance, AI can analyze single-cell data to identify cell subpopulations sensitive to chemotherapy or targeted therapy, customizing compounds or treatment methods (23, 85, 86). Combining single-cell technology and AI can also analyze treatment failure mechanisms, identifying cell subpopulation evolution patterns and underlying molecular mechanisms to develop strategies overcoming treatment resistance (8789). Gene editing, combined with precise molecular targets from single-cell analysis, offers new treatment strategies by editing specific genes in certain cell subpopulations (90, 91).However, integrating single-cell technology, AI, and gene editing faces challenges, including technical complexity, ethical issues, data processing, and clinical application aspects. Ensuring data security and patient privacy is crucial when handling sensitive medical data (9294). Technological advancements may exacerbate medical resource disparities, necessitating international cooperation and policy-making to improve access to advanced technologies in low-income countries (95, 96). Ethical considerations include the implications of applying gene editing technologies like CRISPR-Cas9 to human applications, which have sparked widespread ethical debates. A study in Nature (2022) highlighted potential ethical concerns with gene editing, including effects on offspring, off-target effects, and social equity issues (97). Ensuring the safe and ethical use of these advanced technologies remains a significant challenge that must be addressed.

7 Discussion

Single-cell technologies have fundamentally transformed our comprehension of the tumor microenvironment (TME) in pancreatic cancer, uncovering significant heterogeneity not just among different tumors but also within the same tumor. This detailed analysis has illuminated the complex interplay between various immune cell types, such as T cells, macrophages, and MDSCs, within the TME, each playing distinct roles in tumor immunity (71). Through identifying specific subpopulations of immune cells that either support or hinder the anti-tumor immune response, such as specific T cell exhaustion profiles and myeloid cell markers, these technologies provide insights that could lead to the development of novel therapeutic strategies aimed at enhancing immunotherapy efficacy (98).

Exploring single-cell mechanisms offers unparalleled opportunities to optimize therapeutic strategies in pancreatic cancer. By elucidating the molecular signals and pathways that control the behavior of individual cell types within the TME, researchers can develop targeted therapies to alter the immune landscape in favor of an effective anti-tumor response. This includes strategies like targeting the PD-1/PD-L1 axis in T cells, which, despite limited efficacy, remains promising (99). Single-cell analyses not only shed light on resistance mechanisms but also guide the integration of immunotherapies with other treatments (67, 100), such as chemotherapy, aiming to surmount existing therapeutic barriers. Furthermore, these analyses hold the potential for personalizing medicine by identifying biomarkers that predict a patient’s response to immunotherapy, thereby tailoring treatments to maximize efficacy and minimize the risk of ineffective treatment exposure (101).

However, it is crucial to acknowledge the current limitations of single-cell technology and their impact on clinical applications. One significant limitation is the technical complexity and high cost associated with single-cell sequencing, which can limit its widespread adoption in clinical settings, particularly in low-resource environments. High-throughput single-cell analyses require sophisticated equipment, substantial computational resources, and specialized expertise, posing accessibility challenges (102). Additionally, the vast and complex data generated from single-cell technologies necessitate advanced computational methods and bioinformatics tools for proper analysis and interpretation, leading to challenges in data integration and standardization essential for translating single-cell insights into clinically actionable information (103). Variability in sample preparation and sequencing protocols can introduce biases, affecting reproducibility and reliability of results (102). Another limitation is the potential for incomplete cellular profiling, where certain cell types or states may be underrepresented or missed entirely, leading to gaps in understanding the TME, especially when identifying rare cell populations crucial for tumor progression and treatment response (104).

Despite these limitations, integrating single-cell technologies with advanced computational methods and machine learning algorithms promises to enhance our understanding of the TME, predict treatment outcomes more accurately, and uncover novel therapeutic targets (25). Interdisciplinary collaboration, uniting biologists, clinicians, and data scientists, is pivotal for translating complex biological insights into actionable, clinically viable treatments. Single-cell technologies thus stand as an indispensable tool in the quest to improve pancreatic cancer patient outcomes through more effective, personalized immunotherapeutic strategies, marking a significant step forward in the fight against this challenging disease (105).

Author contributions

RW: Conceptualization, Data curation, Writing – original draft, Writing – review & editing. JL: Writing – original draft, Writing – review & editing. BJ: Writing – original draft, Writing – review & editing. BG: Writing – original draft, Writing – review & editing. HHL: Writing – original draft, Writing – review & editing. FY: Writing – original draft, Writing – review & editing. YY: Writing – original draft, Writing – review & editing. ZC: Writing – original draft, Writing – review & editing. HL: Writing – original draft, Writing – review & editing. CC: Writing – original draft, Writing – review & editing. KX: Writing – original draft, Writing – review & editing. BL: Writing – original draft, Writing – review & editing. XY: Supervision, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by the Science & Technology Department of Sichuan Province (No.: 2023YFQ0101)and the Luzhou Science and Technology Bureau (No: 2022YFS0625-B3).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Wang S, Zheng Y, Yang F, Zhu L, Zhu X-Q, Wang Z-F, et al. The molecular biology of pancreatic adenocarcinoma: translational challenges and clinical perspectives. Signal Transduct Target Ther. (2021) 6:249. doi: 10.1038/s41392-021-00659-4

PubMed Abstract | Crossref Full Text | Google Scholar

2. Sohal DPS, Kennedy EB, Khorana A, Copur MS, Crane CH, Garrido-Laguna I, et al. Metastatic pancreatic cancer: ASCO clinical practice guideline update. J Clin Oncology: Off J Am Soc Clin Oncol. (2018) 36:2545–56. doi: 10.1200/jco.2018.78.9636

Crossref Full Text | Google Scholar

3. Lambert A, Jarlier M, Gourgou Bourgade S, Conroy T. Response to FOLFIRINOX by gender in patients with metastatic pancreatic cancer: Results from the PRODIGE 4/ ACCORD 11 randomized trial. PLoS One. (2017) 12:e0183288. doi: 10.1371/journal.pone.0183288

PubMed Abstract | Crossref Full Text | Google Scholar

4. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. New Engl J Med. (2013) 369:1691–703. doi: 10.1056/nejmoa1304369

PubMed Abstract | Crossref Full Text | Google Scholar

5. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. (2022) 72:7-33. doi: 10.3322/caac.21708

PubMed Abstract | Crossref Full Text | Google Scholar

6. Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet. (2020) 395:2008–20. doi: 10.1016/S0140-6736(20)30974-0

PubMed Abstract | Crossref Full Text | Google Scholar

7. Kanno A, Masamune A, Hanada K, Maguchi H, Shimizu Y, Ueki T, et al. Multicenter study of early pancreatic cancer in Japan. Pancreatology. (2018) 18:61–7. doi: 10.1016/j.pan.2017.11.007

PubMed Abstract | Crossref Full Text | Google Scholar

8. Blackford AL, Canto MI, Klein AP, Hruban RH, Goggins M. Recent trends in the incidence and survival of stage 1A pancreatic cancer: A surveillance, epidemiology, and end results analysis. J Natl Cancer Inst. (2020) 112:1162–9. doi: 10.1093/jnci/djaa004

PubMed Abstract | Crossref Full Text | Google Scholar

9. Kaur S, Baine MJ, Jain M, Sasson AR, Batra SK. Early diagnosis of pancreatic cancer: challenges and new developments. biomark Med. (2012) 6:597–612. doi: 10.2217/bmm.12.69

PubMed Abstract | Crossref Full Text | Google Scholar

10. Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. Nat Rev Dis Primers. (2018) 4:17105. doi: 10.1038/nrdp.2017.105

PubMed Abstract | Crossref Full Text | Google Scholar

11. Esfahani K, Roudaia L, Buhlaiga N, Del Rincon SV, Papneja N, Miller WH. A review of cancer immunotherapy: from the past, to the present, to the future. Curr Oncol. (2020) 27:S87–97. doi: 10.3747/co.27.5223

PubMed Abstract | Crossref Full Text | Google Scholar

12. Wang S-J, Khullar K, Kim S, Yegya-Raman N, Malhotra J, Groisberg R, et al. Effect of cyclo-oxygenase inhibitor use during checkpoint blockade immunotherapy in patients with metastatic melanoma and non-small cell lung cancer. J Immunother Cancer. (2020) 8. doi: 10.1136/jitc-2020-000889

PubMed Abstract | Crossref Full Text | Google Scholar

13. Di Federico A, Mosca M, Pagani R, Carloni R, Frega G, De Giglio A, et al. Immunotherapy in pancreatic cancer: why do we keep failing? A focus on tumor immune microenvironment, predictive biomarkers and treatment outcomes. Cancers (Basel). (2022) 14:2429. doi: 10.3390/cancers14102429

PubMed Abstract | Crossref Full Text | Google Scholar

14. Gupta VK, Sharma NS, Durden B, Garrido VT, Kesh K, Edwards D, et al. Hypoxia-driven oncometabolite L-2HG maintains stemness-differentiation balance and facilitates immune evasion in pancreatic cancer. Cancer Res. (2021) 81:4001–13. doi: 10.1158/0008-5472.can-20-2562

PubMed Abstract | Crossref Full Text | Google Scholar

15. Mucciolo G, Araos Henríquez J, Jihad M, Pinto Teles S, Manansala JS, Li W, et al. EGFR-activated myofibroblasts promote metastasis of pancreatic cancer. Cancer Cell. (2024) 42:101–18. doi: 10.1016/j.ccell.2023.12.002

PubMed Abstract | Crossref Full Text | Google Scholar

16. Kerzel T, Giacca G, Beretta S, Bresesti C, Notaro M, Scotti GM, et al. In vivo macrophage engineering reshapes the tumor microenvironment leading to eradication of liver metastases. Cancer Cell. (2023) 41:1892–910. doi: 10.1016/j.ccell.2023.09.014

PubMed Abstract | Crossref Full Text | Google Scholar

17. Waddell N, Pajic M, Patch A-M, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. (2015) 518:495–501. doi: 10.1038/nature14169

PubMed Abstract | Crossref Full Text | Google Scholar

18. Zhang Y, Lazarus J, Steele NG, Yan W, Lee H-J, Nwosu ZC, et al. Regulatory T-cell depletion alters the tumor microenvironment and accelerates pancreatic carcinogenesis. Cancer Discovery. (2020) 10:422–39. doi: 10.1158/2159-8290.cd-19-0958

PubMed Abstract | Crossref Full Text | Google Scholar

19. Daunke T, Beckinger S, Rahn S, Krüger S, Heckl S, Schäfer H, et al. Expression and role of the immune checkpoint regulator PD-L1 in the tumor-stroma interplay of pancreatic ductal adenocarcinoma. Front Immunol. (2023) 14:1157397. doi: 10.3389/fimmu.2023.1157397

PubMed Abstract | Crossref Full Text | Google Scholar

20. Deng Y, Xia X, Zhao Y, Zhao Z, Martinez C, Yin W, et al. Glucocorticoid receptor regulates PD-L1 and MHC-I in pancreatic cancer cells to promote immune evasion and immunotherapy resistance. Nat Commun. (2021) 12:7041. doi: 10.1038/s41467-021-27349-7

PubMed Abstract | Crossref Full Text | Google Scholar

21. Cappellesso F, Orban M-P, Shirgaonkar N, Berardi E, Serneels J, Neveu M-A, et al. Targeting the bicarbonate transporter SLC4A4 overcomes immunosuppression and immunotherapy resistance in pancreatic cancer. Nat Cancer. (2022) 3:1464–83. doi: 10.1038/s43018-022-00470-2

PubMed Abstract | Crossref Full Text | Google Scholar

22. Padrón LJ, Maurer DM, O'Hara MH, O'Reilly EM, Wolff RA, Wainberg ZA, et al. Sotigalimab and/or nivolumab with chemotherapy in first-line metastatic pancreatic cancer: clinical and immunologic analyses from the randomized phase 2 PRINCE trial. Nat Med. (2022) 28:1167–77. doi: 10.1038/s41591-022-01829-9

PubMed Abstract | Crossref Full Text | Google Scholar

23. Chi H, Su L, Yan Y, Gu X, Su K, Li H, et al. Illuminating the immunological landscape: mitochondrial gene defects in pancreatic cancer through a multiomics lens. Front Immunol. (2024) 15:1375143. doi: 10.3389/fimmu.2024.1375143

PubMed Abstract | Crossref Full Text | Google Scholar

24. Lin W, Noel P, Borazanci EH, Lee J, Amini A, Han IW, et al. Single-cell transcriptome analysis of tumor and stromal compartments of pancreatic ductal adenocarcinoma primary tumors and metastatic lesions. Genome Med. (2020) 12:80. doi: 10.1186/s13073-020-00776-9

PubMed Abstract | Crossref Full Text | Google Scholar

25. Bärthel S, Falcomatà C, Rad R, Theis FJ, Saur D. Single-cell profiling to explore pancreatic cancer heterogeneity, plasticity and response to therapy. Nat Cancer. (2023) 4:454–67. doi: 10.1038/s43018-023-00526-x

PubMed Abstract | Crossref Full Text | Google Scholar

26. Evan T, Wang VM-Y, Behrens A. The roles of intratumour heterogeneity in the biology and treatment of pancreatic ductal adenocarcinoma. Oncogene. (2022) 41:4686–95. doi: 10.1038/s41388-022-02448-x

PubMed Abstract | Crossref Full Text | Google Scholar

27. Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discovery. (2019) 9:1102–23. doi: 10.1158/2159-8290.cd-19-0094

PubMed Abstract | Crossref Full Text | Google Scholar

28. Wang X, Miao J, Wang S, Shen R, Zhang S, Tian Y, et al. Single-cell RNA-seq reveals the genesis and heterogeneity of tumor microenvironment in pancreatic undifferentiated carcinoma with osteoclast-like giant-cells. Mol Cancer. (2022) 21:133. doi: 10.1186/s12943-022-01596-8

PubMed Abstract | Crossref Full Text | Google Scholar

29. Yang K, Yang T, Yu J, Li F, Zhao X. Integrated transcriptional analysis reveals macrophage heterogeneity and macrophage-tumor cell interactions in the progression of pancreatic ductal adenocarcinoma. BMC Cancer. (2023) 23:199. doi: 10.1186/s12885-023-10675-y

PubMed Abstract | Crossref Full Text | Google Scholar

30. Zheng L, Qin S, Si W, Wang A, Xing B, Gao R, et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science. (2021) 374:abe6474. doi: 10.1126/science.abe6474

PubMed Abstract | Crossref Full Text | Google Scholar

31. Hu J, Zhang L, Xia H, Yan Y, Zhu X, Sun F, et al. Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med. (2023) 15:14. doi: 10.1186/s13073-023-01164-9

PubMed Abstract | Crossref Full Text | Google Scholar

32. Li S, Mirlekar B, Johnson BM, Brickey WJ, Wrobel JA, Yang N, et al. STING-induced regulatory B cells compromise NK function in cancer immunity. Nature. (2022) 610:373–80. doi: 10.1038/s41586-022-05254-3

PubMed Abstract | Crossref Full Text | Google Scholar

33. Tang R, Liu X, Wang W, Hua J, Xu J, Liang C, et al. Role of tumor mutation burden-related signatures in the prognosis and immune microenvironment of pancreatic ductal adenocarcinoma. Cancer Cell Int. (2021) 21:196. doi: 10.1186/s12935-021-01900-4

PubMed Abstract | Crossref Full Text | Google Scholar

34. Closa D. Pancreatic cancer, stroma, and exosomes. J Physiol Biochem. (2023) 79:205–11. doi: 10.1007/s13105-022-00898-1

PubMed Abstract | Crossref Full Text | Google Scholar

35. Moncada R, Barkley D, Wagner F, Chiodin M, Devlin JC, Baron M, et al. Integrating microarray-based spatial transcriptomics and single-cell RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat Biotechnol. (2020) 38:333–42. doi: 10.1038/s41587-019-0392-8

PubMed Abstract | Crossref Full Text | Google Scholar

36. Pan Y, Lu F, Fei Q, Yu X, Xiong P, Yu X, et al. Single-cell RNA sequencing reveals compartmental remodeling of tumor-infiltrating immune cells induced by anti-CD47 targeting in pancreatic cancer. J Hematol Oncol. (2019) 12:124. doi: 10.1186/s13045-019-0822-6

PubMed Abstract | Crossref Full Text | Google Scholar

37. Hou S, Zhao Y, Chen J, Lin Y, Qi X. Tumor-associated macrophages in colorectal cancer metastasis: molecular insights and translational perspectives. J Transl Med. (2024) 22:62. doi: 10.1186/s12967-024-04856-x

PubMed Abstract | Crossref Full Text | Google Scholar

38. Kartikasari AER, Huertas CS, Mitchell A, Plebanski M. Tumor-induced inflammatory cytokines and the emerging diagnostic devices for cancer detection and prognosis. Front Oncol. (2021) 11:692142. doi: 10.3389/fonc.2021.692142

PubMed Abstract | Crossref Full Text | Google Scholar

39. Caligola S, De Sanctis F, Canè S, Ugel S. Breaking the immune complexity of the tumor microenvironment using single-cell technologies. Front Genet. (2022) 13:867880. doi: 10.3389/fgene.2022.867880

PubMed Abstract | Crossref Full Text | Google Scholar

40. McRitchie BR, Akkaya B. Exhaust the exhausters: Targeting regulatory T cells in the tumor microenvironment. Front Immunol. (2022) 13:940052. doi: 10.3389/fimmu.2022.940052

PubMed Abstract | Crossref Full Text | Google Scholar

41. Rana M, Kansal R, Chaib M, Teng B, Morrrison M, Hayes DN, et al. The pancreatic cancer immune tumor microenvironment is negatively remodeled by gemcitabine while TGF-β receptor plus dual checkpoint inhibition maintains antitumor immune cells. Mol Carcinog. (2022) 61:549–57. doi: 10.1002/mc.23401

PubMed Abstract | Crossref Full Text | Google Scholar

42. Zhang K, Chen Y, Zhu J, Ge X, Wu J, Xu P, et al. Advancement of single-cell sequencing for clinical diagnosis and treatment of pancreatic cancer. Front In Med. (2023) 10:1213136. doi: 10.3389/fmed.2023.1213136

Crossref Full Text | Google Scholar

43. Xia Q, Jia J, Hu C, Lu J, Li J, Xu H, et al. Tumor-associated macrophages promote PD-L1 expression in tumor cells by regulating PKM2 nuclear translocation in pancreatic ductal adenocarcinoma. Oncogene. (2022) 41:865–77. doi: 10.1038/s41388-021-02133-5

PubMed Abstract | Crossref Full Text | Google Scholar

44. Huber M, Brehm CU, Gress TM, Buchholz M, Alashkar Alhamwe B, von Strandmann EP, et al. The immune microenvironment in pancreatic cancer. Int J Mol Sci. (2020) 21:7307. 10.3390/ijms21197307

PubMed Abstract | Google Scholar

45. Mota Reyes C, Demir E, Çifcibaşı K, Istvanffy R, Friess H, Demir IE. Regulatory T cells in pancreatic cancer: of mice and men. Cancers. (2022) 14:4582. doi: 10.3390/cancers14194582

PubMed Abstract | Crossref Full Text | Google Scholar

46. Yang S, Liu Q, Liao Q. Tumor-associated macrophages in pancreatic ductal adenocarcinoma: origin, polarization, function, and reprogramming. Front Cell Dev Biol. (2020) 8:607209. doi: 10.3389/fcell.2020.607209

PubMed Abstract | Crossref Full Text | Google Scholar

47. Lin H-J, Liu Y, Caroland K, Lin J. Polarization of cancer-associated macrophages maneuver neoplastic attributes of pancreatic ductal adenocarcinoma. Cancers. (2023) 15:3507. doi: 10.3390/cancers15133507

PubMed Abstract | Crossref Full Text | Google Scholar

48. Yang Q, Guo N, Zhou Y, Chen J, Wei Q, Han M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm Sin B. (2020) 10:2156–70. doi: 10.1016/j.apsb.2020.04.004

PubMed Abstract | Crossref Full Text | Google Scholar

49. Ferrara B, Pignatelli C, Cossutta M, Citro A, Courty J, Piemonti L. The extracellular matrix in pancreatic cancer: description of a complex network and promising therapeutic options. Cancers. (2021) 13:4442. doi: 10.3390/cancers13174442

PubMed Abstract | Crossref Full Text | Google Scholar

50. Carvalho TMA, Di Molfetta D, Greco MR, Koltai T, Alfarouk KO, Reshkin SJ, et al. Tumor microenvironment features and chemoresistance in pancreatic ductal adenocarcinoma: insights into targeting physicochemical barriers and metabolism as therapeutic approaches. Cancers. (2021) 13:6135. doi: 10.3390/cancers13236135

PubMed Abstract | Crossref Full Text | Google Scholar

51. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. (2020) 20:174–86. doi: 10.1038/s41568-019-0238-1

PubMed Abstract | Crossref Full Text | Google Scholar

52. Oh K, Yoo YJ, Torre-Healy LA, Rao M, Fassler D, Wang P, et al. Coordinated single-cell tumor microenvironment dynamics reinforce pancreatic cancer subtype. Nat Commun. (2023) 14:5226. doi: 10.1038/s41467-023-40895-6

PubMed Abstract | Crossref Full Text | Google Scholar

53. Zhu J, Zhang K, Chen Y, Ge X, Wu J, Xu P, et al. Progress of single-cell RNA sequencing combined with spatial transcriptomics in tumour microenvironment and treatment of pancreatic cancer. J Transl Med. (2024) 22:563. doi: 10.1186/s12967-024-05307-3

PubMed Abstract | Crossref Full Text | Google Scholar

54. Werba G, Weissinger D, Kawaler EA, Zhao E, Kalfakakou D, Dhara S, et al. Single-cell RNA sequencing reveals the effects of chemotherapy on human pancreatic adenocarcinoma and its tumor microenvironment. Nat Commun. (2023) 14:797. doi: 10.1038/s41467-023-36296-4

PubMed Abstract | Crossref Full Text | Google Scholar

55. Chen Q, Yin H, Jiang Z, He T, Xie Y, Mao W, et al. Poor clinical outcomes and immunoevasive contexture in CD161+CD8+ T cells barren human pancreatic cancer. J Immunother Cancer. (2024) 12. doi: 10.1136/jitc-2023-008694

Crossref Full Text | Google Scholar

56. Wang H, Chen L, Qi L, Jiang N, Zhang Z, Guo H, et al. A single-cell atlas of tumor-infiltrating immune cells in pancreatic ductal adenocarcinoma. Mol Cell Proteomics: MCP. (2022) 21:100258. doi: 10.1016/j.mcpro.2022.100258

Crossref Full Text | Google Scholar

57. Li Z, Mo F, Guo K, Ren S, Wang Y, Chen Y, et al. Nanodrug-bacteria conjugates-mediated oncogenic collagen depletion enhances immune checkpoint blockade therapy against pancreatic cancer. Med (New York NY). (2024) 5;348–67. doi: 10.1016/j.medj.2024.02.012

Crossref Full Text | Google Scholar

58. Salcher S, Sturm G, Horvath L, Untergasser G, Kuempers C, Fotakis G, et al. High-resolution single-cell atlas reveals diversity and plasticity of tissue-resident neutrophils in non-small cell lung cancer. Cancer Cell. (2022) 40:1503–20. doi: 10.1016/j.ccell.2022.10.008

PubMed Abstract | Crossref Full Text | Google Scholar

59. Qiu X, Shi Z, Tong F, Lu C, Zhu Y, Wang Q, et al. Biomarkers for predicting tumor response to PD-1 inhibitors in patients with advanced pancreatic cancer. Hum Vaccines Immunotherapeutics. (2023) 19:2178791. doi: 10.1080/21645515.2023.2178791

Crossref Full Text | Google Scholar

60. Liu R, Dollinger E, Nie Q. Machine learning of single cell transcriptomic data from anti-PD-1 responders and non-responders reveals distinct resistance mechanisms in skin cancers and PDAC. Front Genet. (2021) 12:806457. doi: 10.3389/fgene.2021.806457

PubMed Abstract | Crossref Full Text | Google Scholar

61. Ma Y, Li J, Wang H, Chiu Y, Kingsley CV, Fry D, et al. Combination of PD-1 inhibitor and OX40 agonist induces tumor rejection and immune memory in mouse models of pancreatic cancer. Gastroenterology. (2020) 159:306–19. doi: 10.1053/j.gastro.2020.03.018

PubMed Abstract | Crossref Full Text | Google Scholar

62. Boutilier AJ, Elsawa SF. Macrophage polarization states in the tumor microenvironment. Int J Mol Sci. (2021) 22:6995. doi: 10.3390/ijms22136995

PubMed Abstract | Crossref Full Text | Google Scholar

63. Tichet M, Wullschleger S, Chryplewicz A, Fournier N, Marcone R, Kauzlaric A, et al. Bispecific PD1-IL2v and anti-PD-L1 break tumor immunity resistance by enhancing stem-like tumor-reactive CD8+ T cells and reprogramming macrophages. Immunity. (2023) 56:162–79. doi: 10.1016/j.immuni.2022.12.006

PubMed Abstract | Crossref Full Text | Google Scholar

64. Park SY, Ter-Saakyan S, Faraci G, Lee HY. Immune cell identifier and classifier (ImmunIC) for single cell transcriptomic readouts. Sci Rep. (2023) 13:12093. doi: 10.1038/s41598-023-39282-4

PubMed Abstract | Crossref Full Text | Google Scholar

65. Gao S, Wu Z, Arnold B, Diamond C, Batchu S, Giudice V, et al. Single-cell RNA sequencing coupled to TCR profiling of large granular lymphocyte leukemia T cells. Nat Commun. (2022) 13:1982. doi: 10.1038/s41467-022-29175-x

PubMed Abstract | Crossref Full Text | Google Scholar

66. Hou X, Hong X, Ou M, Meng S, Wang T, Liao S, et al. Analysis of gene expression and TCR/B cell receptor profiling of immune cells in primary sjögren's syndrome by single-cell sequencing. J Immunol (Baltimore Md: 1950). (2022) 209:238–49. doi: 10.4049/jimmunol.2100803

Crossref Full Text | Google Scholar

67. Khodayari S, Khodayari H, Saeedi E, Mahmoodzadeh H, Sadrkhah A, Nayernia K. Single-cell transcriptomics for unlocking personalized cancer immunotherapy: toward targeting the origin of tumor development immunogenicity. Cancers. (2023) 15:3615. doi: 10.3390/cancers15143615

PubMed Abstract | Crossref Full Text | Google Scholar

68. Montagne JM, Jaffee EM, Fertig EJ. Multiomics empowers predictive pancreatic cancer immunotherapy. J Immunol (Baltimore Md: 1950). (2023) 210:859–68. doi: 10.4049/jimmunol.2200660

Crossref Full Text | Google Scholar

69. Xiong C, Zhu Y, Xue M, Jiang Y, Zhong Y, Jiang L, et al. Tumor-associated macrophages promote pancreatic ductal adenocarcinoma progression by inducing epithelial-to-mesenchymal transition. Aging. (2021) 13:3386–404. doi: 10.18632/aging.202264

PubMed Abstract | Crossref Full Text | Google Scholar

70. Wang S, Wang J, Chen Z, Luo J, Guo W, Sun L, et al. Targeting M2-like tumor-associated macrophages is a potential therapeutic approach to overcome antitumor drug resistance. NPJ Precis Oncol. (2024) 8:31. doi: 10.1038/s41698-024-00522-z

PubMed Abstract | Crossref Full Text | Google Scholar

71. Byeon S, du Toit-Thompson T, Gillson J, Gill AJ, Samra JS, Mittal A, et al. Heterogeneous tumor microenvironment in pancreatic ductal adenocarcinoma: An emerging role of single-cell analysis. Cancer Med. (2023) 12:18020–31. doi: 10.1002/cam4.6407

PubMed Abstract | Crossref Full Text | Google Scholar

72. Chijimatsu R, Kobayashi S, Takeda Y, Kitakaze M, Tatekawa S, Arao Y, et al. Establishment of a reference single-cell RNA sequencing dataset for human pancreatic adenocarcinoma. IScience. (2022) 25:104659. doi: 10.1016/j.isci.2022.104659

PubMed Abstract | Crossref Full Text | Google Scholar

73. Lin X, Zong C, Zhang Z, Fang W, Xu P. Progresses in biomarkers for cancer immunotherapy. MedComm. (2023) 4:e387. doi: 10.1002/mco2.387

PubMed Abstract | Crossref Full Text | Google Scholar

74. Mahadevan KK, McAndrews KM, LeBleu VS, Yang S, Lyu H, Li B, et al. KRASG12D inhibition reprograms the microenvironment of early and advanced pancreatic cancer to promote FAS-mediated killing by CD8+ T cells. Cancer Cell. (2023) 41:1606–20. doi: 10.1016/j.ccell.2023.07.002

PubMed Abstract | Crossref Full Text | Google Scholar

75. Pan Y, Gao J, Lin J, Ma Y, Hou Z, Lin Y, et al. High-dimensional single-cell analysis unveils distinct immune signatures of peripheral blood in patients with pancreatic ductal adenocarcinoma. Front In Endocrinol. (2023) 14:1181538. doi: 10.3389/fendo.2023.1181538

Crossref Full Text | Google Scholar

76. Wang D-R, Wu X-L, Sun Y-L. Therapeutic targets and biomarkers of tumor immunotherapy: response versus non-response. Signal Transduct Target Ther. (2022) 7:331. doi: 10.1038/s41392-022-01136-2

PubMed Abstract | Crossref Full Text | Google Scholar

77. Lu C, Talukder A, Savage NM, Singh N, Liu K. JAK-STAT-mediated chronic inflammation impairs cytotoxic T lymphocyte activation to decrease anti-PD-1 immunotherapy efficacy in pancreatic cancer. Oncoimmunology. (2017) 6:e1291106. doi: 10.1080/2162402x.2017.1291106

PubMed Abstract | Crossref Full Text | Google Scholar

78. Halbrook CJ, Lyssiotis CA, Pasca di Magliano M, Maitra A. Pancreatic cancer: Advances and challenges. Cell. (2023) 186:1729–54. doi: 10.1016/j.cell.2023.02.014

PubMed Abstract | Crossref Full Text | Google Scholar

79. Jiang P, Sinha S, Aldape K, Hannenhalli S, Sahinalp C, Ruppin E. Big data in basic and translational cancer research. Nat Rev Cancer. (2022) 22:625–39. doi: 10.1038/s41568-022-00502-0

PubMed Abstract | Crossref Full Text | Google Scholar

80. Hosein AN, Huang H, Wang Z, Parmar K, Du W, Huang J, et al. Cellular heterogeneity during mouse pancreatic ductal adenocarcinoma progression at single-cell resolution. JCI Insight. (2019) 5:e129212. doi: 10.1172/jci.insight.129212

PubMed Abstract | Crossref Full Text | Google Scholar

81. Zhang S, Fang W, Zhou S, Zhu D, Chen R, Gao X, et al. Single cell transcriptomic analyses implicate an immunosuppressive tumor microenvironment in pancreatic cancer liver metastasis. Nat Commun. (2023) 14:5123. doi: 10.1038/s41467-023-40727-7

PubMed Abstract | Crossref Full Text | Google Scholar

82. Liu ZL, Meng XY, Bao RJ, Shen MY, Sun JJ, Chen WD, et al. Single cell deciphering of progression trajectories of the tumor ecosystem in head and neck cancer. Nat Commun. (2024) 15:2595. doi: 10.1038/s41467-024-46912-6

PubMed Abstract | Crossref Full Text | Google Scholar

83. Busnatu Ș, Niculescu A-G, Bolocan A, Petrescu GED, Păduraru DN, Năstasă I, et al. Clinical applications of artificial intelligence-an updated overview. J Clin Med. (2022) 11:2265. doi: 10.3390/jcm11082265

PubMed Abstract | Crossref Full Text | Google Scholar

84. Sharma A, Lysenko A, Jia S, Boroevich KA, Tsunoda T. Advances in AI and machine learning for predictive medicine. J Hum Genet. (2024) 69:487–97. doi: 10.1038/s10038-024-01231-y

PubMed Abstract | Crossref Full Text | Google Scholar

85. Russo V, Lallo E, Munnia A, Spedicato M, Messerini L, D'Aurizio R, et al. Artificial intelligence predictive models of response to cytotoxic chemotherapy alone or combined to targeted therapy for metastatic colorectal cancer patients: A systematic review and meta-analysis. Cancers. (2022) 14:4012. doi: 10.3390/cancers14164012

PubMed Abstract | Crossref Full Text | Google Scholar

86. Fiste O, Gkiozos I, Charpidou A, Syrigos NK. Artificial intelligence-based treatment decisions: A new era for NSCLC. Cancers. (2024) 16:831. doi: 10.3390/cancers16040831

PubMed Abstract | Crossref Full Text | Google Scholar

87. Wen L, Li G, Huang T, Geng W, Pei H, Yang J, et al. Single-cell technologies: From research to application. Innovation (Cambridge (Mass)). (2022) 3:100342. doi: 10.1016/j.xinn.2022.100342

PubMed Abstract | Crossref Full Text | Google Scholar

88. Morgan D, Tergaonkar V. Unraveling B cell trajectories at single cell resolution. Trends In Immunol. (2022) 43:210–29. doi: 10.1016/j.it.2022.01.003

Crossref Full Text | Google Scholar

89. Huang X, Chi H, Gou S, Guo X, Li L, Peng G, et al. An aggrephagy-related lncRNA signature for the prognosis of pancreatic adenocarcinoma. Genes (Basel). (2023) 14:124. doi: 10.3390/genes14010124

PubMed Abstract | Crossref Full Text | Google Scholar

90. Barragán-Álvarez CP, Flores-Fernandez JM, Hernández-Pérez OR, Ávila-Gónzalez D, Díaz NF, Padilla-Camberos E, et al. Recent advances in the use of CRISPR/Cas for understanding the early development of molecular gaps in glial cells. Front Cell Dev Biol. (2022) 10:947769. doi: 10.3389/fcell.2022.947769

PubMed Abstract | Crossref Full Text | Google Scholar

91. Chi H, Peng G, Wang R, Yang F, Xie X, Zhang J, et al. Cuprotosis programmed-cell-death-related lncRNA signature predicts prognosis and immune landscape in PAAD patients. Cells. (2022) 11:3436. doi: 10.3390/cells11213436

PubMed Abstract | Crossref Full Text | Google Scholar

92. Blise KE, Sivagnanam S, Betts CB, Betre K, Kirchberger N, Tate BJ, et al. Machine learning links T-cell function and spatial localization to neoadjuvant immunotherapy and clinical outcome in pancreatic cancer. Cancer Immunol Res. (2024) 12:544–58. doi: 10.1158/2326-6066.cir-23-0873

PubMed Abstract | Crossref Full Text | Google Scholar

93. Tan CL, Lindner K, Boschert T, Meng Z, Rodriguez Ehrenfried A, De Roia A, et al. Prediction of tumor-reactive T cell receptors from scRNA-seq data for personalized T cell therapy. Nat Biotechnol. (2024). doi: 10.1038/s41587-024-02161-y

PubMed Abstract | Crossref Full Text | Google Scholar

94. Daher H, Punchayil SA, Ismail AAE, Fernandes RR, Jacob J, Algazzar MH, et al. Advancements in pancreatic cancer detection: integrating biomarkers, imaging technologies, and machine learning for early diagnosis. Cureus. (2024) 16:e56583. doi: 10.7759/cureus.56583

PubMed Abstract | Crossref Full Text | Google Scholar

95. Rocher L, Hendrickx JM, de Montjoye Y-A. Estimating the success of re-identifications in incomplete datasets using generative models. Nat Commun. (2019) 10:3069. doi: 10.1038/s41467-019-10933-3

PubMed Abstract | Crossref Full Text | Google Scholar

96. Romanello M, Di Napoli C, Drummond P, Green C, Kennard H, Lampard P, et al. The 2022 report of the Lancet Countdown on health and climate change: health at the mercy of fossil fuels. Lancet. (2022) 400:1619–54. doi: 10.1016/S0140-6736(22)02314-5

PubMed Abstract | Crossref Full Text | Google Scholar

97. Almeida M, Ranisch R. Beyond safety: mapping the ethical debate on heritable genome editing interventions. Humanities Soc Sci Commun. (2022) 9:139. doi: 10.1057/s41599-022-01147-y

Crossref Full Text | Google Scholar

98. Cheng X, Wang H, Wang Z, Zhu B, Long H. Tumor-associated myeloid cells in cancer immunotherapy. J Hematol Oncol. (2023) 16:71. doi: 10.1186/s13045-023-01473-x

PubMed Abstract | Crossref Full Text | Google Scholar

99. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res. (2020) 10:727–42. Available at: https://pubmed.ncbi.nlm.nih.gov/32266087/.

PubMed Abstract | Google Scholar

100. Chen S, Jiang W, Du Y, Yang M, Pan Y, Li H, et al. Single-cell analysis technologies for cancer research: from tumor-specific single cell discovery to cancer therapy. Front Genet. (2023) 14:1276959. doi: 10.3389/fgene.2023.1276959

PubMed Abstract | Crossref Full Text | Google Scholar

101. Tian J, Bai X, Quek C. Single-cell informatics for tumor microenvironment and immunotherapy. Int J Mol Sci. (2024) 25:4485. doi: 10.3390/ijms25084485

PubMed Abstract | Crossref Full Text | Google Scholar

102. Xu Y, Su G-H, Ma D, Xiao Y, Shao Z-M, Jiang Y-Z. Technological advances in cancer immunity: from immunogenomics to single-cell analysis and artificial intelligence. Signal Transduct Target Ther. (2021) 6:312. doi: 10.1038/s41392-021-00729-7

PubMed Abstract | Crossref Full Text | Google Scholar

103. Chen J, Wang X, Ma A, Wang Q-E, Liu B, Li L, et al. Deep transfer learning of cancer drug responses by integrating bulk and single-cell RNA-seq data. Nat Commun. (2022) 13:6494. doi: 10.1038/s41467-022-34277-7

PubMed Abstract | Crossref Full Text | Google Scholar

104. Sorin M, Rezanejad M, Karimi E, Fiset B, Desharnais L, Perus LJM, et al. Single-cell spatial landscapes of the lung tumour immune microenvironment. Nature. (2023) 614:548–54. doi: 10.1038/s41586-022-05672-3

PubMed Abstract | Crossref Full Text | Google Scholar

105. Yousuf S, Qiu M, Voith von Voithenberg L, Hulkkonen J, Macinkovic I, Schulz AR, et al. Spatially resolved multi-omics single-cell analyses inform mechanisms of immune dysfunction in pancreatic cancer. Gastroenterology. (2023) 165:891–908. doi: 10.1053/j.gastro.2023.05.036

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: pancreatic cancer, immunotherapy, single-cell technology, tumor microenvironment, predictive biomarkers, personalized treatment

Citation: Wang R, Liu J, Jiang B, Gao B, Luo H, Yang F, Ye Y, Chen Z, Liu H, Cui C, Xu K, Li B and Yang X (2024) A single-cell perspective on immunotherapy for pancreatic cancer: from microenvironment analysis to therapeutic strategy innovation. Front. Immunol. 15:1454833. doi: 10.3389/fimmu.2024.1454833

Received: 25 June 2024; Accepted: 08 October 2024;
Published: 30 October 2024.

Edited by:

Raquel Alarcon Rodriguez, University of Almeria, Spain

Reviewed by:

Yuquan Chen, Monash University, Australia

Copyright © 2024 Wang, Liu, Jiang, Gao, Luo, Yang, Ye, Chen, Liu, Cui, Xu, Li and Yang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ke Xu, Y3FnaHh1a2VAY3F1LmVkdS5jbg==; Bo Li, bGlib2VyMjAwMkAxMjYuY29t; Xiaoli Yang, eWFuZ3hpYW9saUBzd211LmVkdS5jbg==

These authors have contributed equally to this work.

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.