Skip to main content
Erschienen in:

Open Access 18.12.2024 | short review

Personalized decision making of neoadjuvant chemotherapy vs. upfront surgery in pancreatic cancer by a simple blood collection?

verfasst von: Patrick Kirchweger, MD, PhD, Bernhard Doleschal, MD, PhD, Holger Rumpold, MD, PD, Helwig Wundsam, MD, PD, Matthias Biebl, MD, Prof.

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 1/2025

Summary

Pancreatic cancer still has dismal survival rates and high rates of early recurrence despite improvements of multimodal treatment options and more and more aggressive surgical approaches in recent years. Thus, precise and personalized management strategies to improve patient outcomes are needed. Circulating tumor DNA (ctDNA), a component of cell-free DNA (cfDNA) in body fluids, harbors genetic and epigenetic signatures of tumors and can be detected noninvasively for example through simple blood collections or peritoneal fluid during staging laparoscopy (liquid biopsies). This biomarker provides real-time insights into systemic tumor burden, heterogeneity, and genetic profile and has been proven to be of significant prognostic relevance for several gastrointestinal malignancies. Furthermore, the testing of ctDNA has emerged as a pivotal prognostic biomarker to indicate patients with high biological risk for recurrence and worse overall survival. Especially in pancreatic cancer, it has been shown that preoperative ctDNA detectability in peripheral blood is associated with systemic tumor burden (even volumetric). This indicates potential micrometastatic or subclinical disseminated disease, suggesting a benefit from neoadjuvant chemotherapy to address the systemic component of the disease prior to surgery. Furthermore, dynamic changes in ctDNA during systemic treatment can predict therapeutic response and guide adjustments in treatment regimens. Postoperatively, ctDNA presence could assist in detecting minimal residual disease which also predicts early relapse, facilitating timely intervention (or immediate adjuvant chemotherapy as already shown in colorectal cancer, e.g., Dynamic trial). Therefore, perioperative ctDNA detection has the potential to refine the management of pancreatic cancer, enhancing decision-making processes and optimizing personalized treatment approaches in serving as an additional highly sensitive biomarker for guiding treatment decisions between upfront surgery and neoadjuvant chemotherapy in patients with resectable pancreatic cancer. The first ever personalized change of treatment decision from upfront surgery (by current gold standard staging) to neoadjuvant chemotherapy based on additional liquid biopsy results was documented in March 2024 (Linz, Austria).
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
cfDNA
Cell-free DNA
CT
Computed tomography
ctDNA
Circulating tumor DNA
CTX
Chemotherapy
DFS
Disease-free survival
LAPC
Locally advanced pancreatic cancer
NCCN
National Comprehensive Cancer Network
NGS
Next generation sequencing
OS
Overall survival
PDAC
Pancreatic ductal adenocarcinoma

Introduction

Being already the third leading cause for cancer-related death in Europe and fourth leading in the world, pancreatic cancer remains one of the most challenging malignancies to treat, facing mere 5‑year survival rates of less than 32% (Ia) to 0.5% (IV) depending on the tumor stage [1, 2]. Although surgical removal is regarded the only curative approach, median overall survival from dissemination on does not exceed 1 year and 80% of patients will experience relapse within 2 years [1, 3]. The management of resectable pancreatic cancer, particularly the decision between upfront surgery versus neoadjuvant chemotherapy, is pivotal and can significantly impact outcomes [4]. Despite current gold standard staging (computed tomography, CA 19‑9 and tissue biopsy), some patients suffer from early recurrence or even initial metastatic stage of the disease explored during explorative laparotomy when intending curative resection (10–15% of patients, 21.3% in locally advanced stage) [5, 6]. The identification of those patients at high biological risk for recurrence or subclinical dissemination is of great interest for better stratification of either systemic or local therapy but is not sufficiently viable with current staging modalities.
Thus, circulating tumor DNA (ctDNA) has emerged as a promising marker indicating tumor burden with high sensitivity in several tumor entities but could be of special value for treatment decision-making in pancreatic cancer due to the relatively easy clinical implementation (small mutational spectrum, low cost) and high prognostic impact, especially in patients with potentially curative resectable pancreatic cancer.

Circulating tumor DNA

Circulating tumor DNA (ctDNA), fragments of DNA shed by tumors into the bloodstream, has emerged as a promising biomarker in several cancers, offering a noninvasive means to provide information for treatment decisions via liquid biopsies. ctDNA represents a fraction of the free DNA found in blood and carries genetic and epigenetic alterations specific to tumors. Its detection and quantification can provide real-time insights into tumor genetics, burden, and heterogeneity, which are invaluable in the precision medicine landscape [7, 8].

ctDNA derivation, prognostic impact, and limitations of CA19-9

In pancreatic cancer, ctDNA is detectable in a significant proportion of patients (10–65% depending on the tumor stage) and correlates with disease burden and prognosis [9]. Studies have shown that higher levels of ctDNA are associated with advanced disease and worse outcomes, suggesting its potential role in staging and prognosis and even outperforms the current gold standard tumor marker CA19‑9 in doing so [4, 1012]. Moreover, several limitations regarding CA19‑9 have to be taken into account. First, its secretion is connected to Lewis antigen and, thus, CA19‑9 cannot be produced in 10–15% of patients (5–10% in Caucasians) [13]. Second, several nonmalignant conditions (e.g., chronic pancreatitis, liver cirrhosis, cholangitis or jaundice which is especially common in patients with pancreatic head tumors [the most common tumor location]) can lead to false elevated CA19-9 levels hampering exact diagnosis [13]. This is especially alarming when considering CA19‑9 as the only biomarker in pancreatic cancer care used for neoadjuvant chemotherapy recommendation when < 500 U/ml acknowledged by NCCN (National Comprehensive Cancer Network) [14, 15]. Generally, making such a fundamental treatment decision solely based on this threshold of < 500 U/ml in addition to the remaining ABC (anatomy: resectable, borderline resectable, locally advanced; biomarker: CA19‑9; C: performance status) factors for clinical staging makes no sense when considering the broad variety of values given for CA19‑9 in one and the same patient depending on the manufacturer (Abott Alinity/Architect +77%, Roche Cobas −48% compared to the consensus mean) [16, 17]. Ultimately, the use of CA19‑9 as screening tool is also hampered by its limited positive predictive value [2].

Neoadjuvant chemotherapy vs. upfront surgery

The traditional approach for resectable pancreatic cancer involves upfront surgery followed by adjuvant chemotherapy [2]. However, this strategy often leads to suboptimal outcomes due to undetected micrometastatic disease at the time of surgery [15]. Neoadjuvant chemotherapy (NACT), on the other hand, can treat micrometastatic disease early, potentially increasing resectability and survival rates. However, consistent data and guidelines (e.g., ESMO, NCCN) suggests NACT for borderline or locally advanced patients but improved outcome for primarily resectable patients undergoing upfront resection instead of NACT [1820].
Role of ctDNA in assessing tumor burden and metastatic potential:
ctDNA levels can serve as a proxy for tumor burden [9]. High preoperative ctDNA levels have been associated with poor surgical outcomes and higher recurrence rates [21]. Thus, detecting ctDNA preoperatively could help identify patients who may benefit more from initial systemic chemotherapy rather than immediate surgery [22]. This has not been proven yet.
Predicting response to neoadjuvant chemotherapy:
Monitoring ctDNA levels during neoadjuvant treatment provides insights into the tumor’s response to therapy [22]. A decrease in ctDNA levels has been correlated with a better pathological response in nonmetastatic PDAC (pancreatic ductal adenocarcinoma) and dynamic changes of ctDNA levels during systemic treatment were associated with early evaluation of response to treatment at metastatic stage [10, 22]. Thus, ctDNA could identify patients who are responding well to chemotherapy before surgery.
Detection of minimal residual disease and early relapse:
Postoperative ctDNA analysis can detect minimal residual disease, offering an early indication of relapse [11]. Patients with detectable ctDNA postsurgery have a higher risk of recurrence and could be candidates for additional or intensified adjuvant therapy [11, 12, 23].

Challenges and limitations

Despite its potential and advances in recent years, clinical implementation of ctDNA still faces several challenges:
  • Sensitivity and specificity vary widely among assays, impacting reliability of the many different approaches available [24].
  • Standardization of ctDNA sampling, processing, and analysis is needed [8].
  • The cost and accessibility of ctDNA testing are concerns that must be addressed to make this a viable option across various healthcare settings [9]. Thus, NGS is neither cost effective nor necessary but used in many studies. Small spectrum ddPCR for the most common KRAS alterations should be sufficient [10].
  • Despite its promising utility, ctDNA testing faces challenges such as variability in assay sensitivity and specificity, a lack of standardization in sample processing, and issues related to cost and accessibility [24]. Ongoing research and technological advancements are addressing these barriers, aiming to integrate ctDNA analysis more effectively into clinical practice [24].

Future directions

Ongoing trials and studies aim to refine the applications of ctDNA in treatment decision-making [12]. Enhanced digital PCR and next-generation sequencing technologies are improving the detection and quantification of ctDNA, paving the way for its routine use in clinical practice [25].

Conclusion

Although the current clinical application is purely experimental, ctDNA analysis bears promising potential to improve the management of resectable pancreatic cancer by providing a noninvasive tool to guide the choice between neoadjuvant chemotherapy and upfront surgery in the future by distinguishing between localized or systemic/advanced disease. This approach is currently subject of clinical trials, but as evidence accumulates, it becomes clear that integrating ctDNA into clinical workflows could enhance the precision of cancer care, offering patients tailored and potentially more effective treatment pathways.
Take-home message
The role of ctDNA in determining the optimal treatment strategies for resectable pancreatic cancer has not been definitively clarified yet but showed promising results in recent studies emphasizing its potential to tailor decisions between neoadjuvant chemotherapy and upfront surgery based on liquid biopsy results in the future.
  • ctDNA displays the actual systemic tumor burden and is assessable with simple blood collection. No prior tissue testing is necessary for prognostic information in pancreatic cancer due to the small mutational spectrum. This allows fast decision making.
  • Both preoperative and postoperative ctDNA detectability in localized pancreatic cancer is associated with worse overall survival and disease-free survival. However, there is no universal testing strategy or cut-off within the literature. Nevertheless, ddPCR at three positive droplets seems very suitable.
  • Pretherapeutic ctDNA detectability could be used to identify patients at high biological risk for early recurrence despite insignificant CT and CA 19‑9 in the future.
  • Prospective interventional trials are needed to establish the clinical applicability and impact of ctDNA-guided treatment in pancreatic cancer; to date, no such trials have been published. The LIQUIPANC trial (NCT06391892), initiated in January 2024 at the Ordensklinikum Linz, Austria, explores a pioneering approach using personalized ctDNA analysis to support treatment decisions for pancreatic cancer. This trial specifically assesses the feasibility of using preoperative ctDNA to guide the choice between chemotherapy and upfront surgery in resectable PDAC. Although data from previous observational studies at this and other centers suggest a potential shift towards more personalized treatment protocols, the broader impact on standard care practices remains to be confirmed. Ongoing research and multicentric validation by others are needed in the future. Additionally, the LIQUIPANC trial will be expanded to include several centers across Austria in 2025 as planned, trying to adopt an iterative approach to integrating ctDNA testing into clinical routines.
    Final results are expected in 2026/2027.

Conflict of interest

P. Kirchweger, B. Doleschal, H. Rumpold, H. Wundsam and M. Biebl declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Strobel O, Neoptolemos J, Jäger D, Büchler MW. Optimizing the outcomes of pancreatic cancer surgery. Nat Rev Clin Oncol. 2019;16:11–26.CrossRefPubMed Strobel O, Neoptolemos J, Jäger D, Büchler MW. Optimizing the outcomes of pancreatic cancer surgery. Nat Rev Clin Oncol. 2019;16:11–26.CrossRefPubMed
2.
Zurück zum Zitat Bugazia D, Al-Najjar E, Esmail A, Abdelrahim S, Abboud K, Abdelrahim A, et al. Pancreatic ductal adenocarcinoma: the latest on diagnosis, molecular profiling, and systemic treatments. Front Oncol. 2024;14:1386699.CrossRefPubMedPubMedCentral Bugazia D, Al-Najjar E, Esmail A, Abdelrahim S, Abboud K, Abdelrahim A, et al. Pancreatic ductal adenocarcinoma: the latest on diagnosis, molecular profiling, and systemic treatments. Front Oncol. 2024;14:1386699.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Watanabe K, Nakamura T, Kimura Y, Motoya M, Kojima S, Kuraya T, et al. Tumor-informed approach improved ctDNA detection rate in Resected pancreatic cancer. Int J Mol Sci. 2022;23:11521.CrossRefPubMedPubMedCentral Watanabe K, Nakamura T, Kimura Y, Motoya M, Kojima S, Kuraya T, et al. Tumor-informed approach improved ctDNA detection rate in Resected pancreatic cancer. Int J Mol Sci. 2022;23:11521.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Hadano N, Murakami Y, Uemura K, Hashimoto Y, Kondo N, Nakagawa N, et al. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br J Cancer. 2016;115:59–65.CrossRefPubMedPubMedCentral Hadano N, Murakami Y, Uemura K, Hashimoto Y, Kondo N, Nakagawa N, et al. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br J Cancer. 2016;115:59–65.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Theijse RT, Stoop TF, Leenart PD, Lutchman KRD, Erdmann JI, Daams F, et al. Surgery for locally advanced pancreatic cancer following induction chemotherapy: a single-center experience. Ann Surg Oncol. 2024;. Theijse RT, Stoop TF, Leenart PD, Lutchman KRD, Erdmann JI, Daams F, et al. Surgery for locally advanced pancreatic cancer following induction chemotherapy: a single-center experience. Ann Surg Oncol. 2024;.
6.
Zurück zum Zitat Walma M, Maggino L, Smits FJ, Borggreve AS, Daamen LA, Groot VP, et al. The difficulty of detecting occult metastases in patients with potentially Resectable pancreatic cancer: development and external validation of a preoperative prediction model. J Clin Med. 2024;13. Walma M, Maggino L, Smits FJ, Borggreve AS, Daamen LA, Groot VP, et al. The difficulty of detecting occult metastases in patients with potentially Resectable pancreatic cancer: development and external validation of a  preoperative prediction model. J Clin Med. 2024;13.
7.
Zurück zum Zitat Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.CrossRefPubMedPubMedCentral Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Kirchweger P, Wundsam HV, Rumpold H. Circulating tumor DNA for diagnosis, prognosis and treatment of gastrointestinal malignancies. World J Clin Oncol. 2022;13:473–84.CrossRefPubMedPubMedCentral Kirchweger P, Wundsam HV, Rumpold H. Circulating tumor DNA for diagnosis, prognosis and treatment of gastrointestinal malignancies. World J Clin Oncol. 2022;13:473–84.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Kirchweger P, Kupferthaler A, Burghofer J, Webersinke G, Jukic E, Schwendinger S, et al. Circulating tumor DNA correlates with tumor burden and predicts outcome in pancreatic cancer irrespective of tumor stage. Eur J Surg Oncol. 2022;48:1046–53.CrossRefPubMed Kirchweger P, Kupferthaler A, Burghofer J, Webersinke G, Jukic E, Schwendinger S, et al. Circulating tumor DNA correlates with tumor burden and predicts outcome in pancreatic cancer irrespective of tumor stage. Eur J Surg Oncol. 2022;48:1046–53.CrossRefPubMed
10.
Zurück zum Zitat Kirchweger P, Kupferthaler A, Burghofer J, Webersinke G, Jukic E, Schwendinger S, et al. Prediction of response to systemic treatment by kinetics of circulating tumor DNA in metastatic pancreatic cancer. Front Oncol. 2022;12:902177.CrossRefPubMedPubMedCentral Kirchweger P, Kupferthaler A, Burghofer J, Webersinke G, Jukic E, Schwendinger S, et al. Prediction of response to systemic treatment by kinetics of circulating tumor DNA in metastatic pancreatic cancer. Front Oncol. 2022;12:902177.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Lee B, Lipton L, Cohen J, Tie J, Javed AA, Li L, et al. Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann Oncol Off J Eur Soc Med Oncol. 2019;30:1472–8.CrossRef Lee B, Lipton L, Cohen J, Tie J, Javed AA, Li L, et al. Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann Oncol Off J Eur Soc Med Oncol. 2019;30:1472–8.CrossRef
12.
Zurück zum Zitat Eckhoff AM, Kanu E, Fletcher A, Bao M, Aushev VN, Spickard E, et al. Initial report: personalized circulating tumor DNA and survival in patients with Resectable pancreatic cancer. Ann Surg Oncol. 2024;31:1444–6.CrossRefPubMed Eckhoff AM, Kanu E, Fletcher A, Bao M, Aushev VN, Spickard E, et al. Initial report: personalized circulating tumor DNA and survival in patients with Resectable pancreatic cancer. Ann Surg Oncol. 2024;31:1444–6.CrossRefPubMed
13.
Zurück zum Zitat Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol. 2007;33:266–70.CrossRefPubMed Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol. 2007;33:266–70.CrossRefPubMed
14.
Zurück zum Zitat Ushida Y, Inoue Y, Ito H, Oba A, Mise Y, Ono Y, et al. High CA19‑9 level in resectable pancreatic cancer is a potential indication of neoadjuvant treatment. Pancreatology. 2021;21:130–7.CrossRefPubMed Ushida Y, Inoue Y, Ito H, Oba A, Mise Y, Ono Y, et al. High CA19‑9 level in resectable pancreatic cancer is a potential indication of neoadjuvant treatment. Pancreatology. 2021;21:130–7.CrossRefPubMed
15.
Zurück zum Zitat Crippa S, Malleo G, Mazzaferro V, Langella S, Ricci C, Casciani F, et al. Futility of up-front resection for anatomically Resectable pancreatic cancer. JAMA Surg. 2024;. Crippa S, Malleo G, Mazzaferro V, Langella S, Ricci C, Casciani F, et al. Futility of up-front resection for anatomically Resectable pancreatic cancer. JAMA Surg. 2024;.
16.
Zurück zum Zitat van Rossum HH, Holdenrieder S, Ballieux BEPB, Badrick TC, Yun Y‑M, Zhang C, et al. Investigating the current harmonization status of tumor markers using global external quality assessment programs: a feasibility study. Clin Chem. 2024;70:669–79.CrossRefPubMed van Rossum HH, Holdenrieder S, Ballieux BEPB, Badrick TC, Yun Y‑M, Zhang C, et al. Investigating the current harmonization status of tumor markers using global external quality assessment programs: a feasibility study. Clin Chem. 2024;70:669–79.CrossRefPubMed
17.
Zurück zum Zitat Dekker EN, van Dam JL, Janssen QP, Besselink MG, DeSilva A, Doppenberg D, et al. Improved clinical staging system for localized pancreatic cancer using the ABC factors: a TAPS consortium study. J Clin Oncol. 2024;42:1357–67.CrossRefPubMed Dekker EN, van Dam JL, Janssen QP, Besselink MG, DeSilva A, Doppenberg D, et al. Improved clinical staging system for localized pancreatic cancer using the ABC factors: a TAPS consortium study. J Clin Oncol. 2024;42:1357–67.CrossRefPubMed
18.
Zurück zum Zitat Ghaneh P, Palmer D, Cicconi S, Jackson R, Halloran CM, Rawcliffe C, et al. Immediate surgery compared with short-course neoadjuvant gemcitabine plus capecitabine, FOLFIRINOX, or chemoradiotherapy in patients with borderline resectable pancreatic cancer (ESPAC5): a four-arm, multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2023;8:157–68.CrossRefPubMed Ghaneh P, Palmer D, Cicconi S, Jackson R, Halloran CM, Rawcliffe C, et al. Immediate surgery compared with short-course neoadjuvant gemcitabine plus capecitabine, FOLFIRINOX, or chemoradiotherapy in patients with borderline resectable pancreatic cancer (ESPAC5): a four-arm, multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2023;8:157–68.CrossRefPubMed
19.
Zurück zum Zitat Springfeld C, Ferrone CR, Katz MHG, Philip PA, Hong TS, Hackert T, et al. Neoadjuvant therapy for pancreatic cancer. Nat Rev Clin Oncol. 2023;20:318–37.CrossRefPubMed Springfeld C, Ferrone CR, Katz MHG, Philip PA, Hong TS, Hackert T, et al. Neoadjuvant therapy for pancreatic cancer. Nat Rev Clin Oncol. 2023;20:318–37.CrossRefPubMed
20.
Zurück zum Zitat Labori KJ, Bratlie SO, Andersson B, Angelsen J‑H, Biörserud C, Björnsson B, et al. Neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer (NORPACT-1): a multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2024;9:205–17.CrossRefPubMed Labori KJ, Bratlie SO, Andersson B, Angelsen J‑H, Biörserud C, Björnsson B, et al. Neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer (NORPACT-1): a multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2024;9:205–17.CrossRefPubMed
21.
Zurück zum Zitat Alqahtani A, Alloghbi A, Coffin P, Yin C, Mukherji R, Weinberg BA. Prognostic utility of preoperative and postoperative KRAS-mutated circulating tumor DNA (ctDNA) in resected pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Surg Oncol. 2023;51:102007.CrossRefPubMed Alqahtani A, Alloghbi A, Coffin P, Yin C, Mukherji R, Weinberg BA. Prognostic utility of preoperative and postoperative KRAS-mutated circulating tumor DNA (ctDNA) in resected pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Surg Oncol. 2023;51:102007.CrossRefPubMed
22.
Zurück zum Zitat Ueberroth BE, Jones JC, Bekaii-Saab TS. Circulating tumor DNA (ctDNA) to evaluate minimal residual disease (MRD), treatment response, and posttreatment prognosis in pancreatic adenocarcinoma. Pancreatology. 2022;22:741–8.CrossRefPubMed Ueberroth BE, Jones JC, Bekaii-Saab TS. Circulating tumor DNA (ctDNA) to evaluate minimal residual disease (MRD), treatment response, and posttreatment prognosis in pancreatic adenocarcinoma. Pancreatology. 2022;22:741–8.CrossRefPubMed
23.
Zurück zum Zitat Cecchini M, Salem RR, Robert M, Czerniak S, Blaha O, Zelterman D, et al. Perioperative modified FOLFIRINOX for Resectable pancreatic cancer: a Nonrandomized controlled trial. JAMA Oncol. 2024; e241575. Cecchini M, Salem RR, Robert M, Czerniak S, Blaha O, Zelterman D, et al. Perioperative modified FOLFIRINOX for Resectable pancreatic cancer: a Nonrandomized controlled trial. JAMA Oncol. 2024; e241575.
24.
Zurück zum Zitat Dao J, Conway PJ, Subramani B, Meyyappan D, Russell S, Mahadevan D. Using cfDNA and ctDNA as oncologic markers: a path to clinical validation. Int J Mol Sci. 2023;24:13219.CrossRefPubMedPubMedCentral Dao J, Conway PJ, Subramani B, Meyyappan D, Russell S, Mahadevan D. Using cfDNA and ctDNA as oncologic markers: a path to clinical validation. Int J Mol Sci. 2023;24:13219.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Lee J‑S, Park SS, Lee YK, Norton JA, Jeffrey SS. Liquid biopsy in pancreatic ductal adenocarcinoma: current status of circulating tumor cells and circulating tumor DNA. Mol Oncol. 2019;13:1623–50.CrossRefPubMedPubMedCentral Lee J‑S, Park SS, Lee YK, Norton JA, Jeffrey SS. Liquid biopsy in pancreatic ductal adenocarcinoma: current status of circulating tumor cells and circulating tumor DNA. Mol Oncol. 2019;13:1623–50.CrossRefPubMedPubMedCentral
Metadaten
Titel
Personalized decision making of neoadjuvant chemotherapy vs. upfront surgery in pancreatic cancer by a simple blood collection?
verfasst von
Patrick Kirchweger, MD, PhD
Bernhard Doleschal, MD, PhD
Holger Rumpold, MD, PD
Helwig Wundsam, MD, PD
Matthias Biebl, MD, Prof.
Publikationsdatum
18.12.2024
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 1/2025
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-024-01013-4