Umeå University's logo

umu.sePublications
Change search
Link to record
Permanent link

Direct link
Publications (10 of 46) Show all publications
Xu, J., Pham, M. D., Corbo, V., Ponz-Sarvise, M., Oni, T., Öhlund, D. & Hwang, C.-I. (2025). Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology. Experimental and Molecular Medicine, 57(1), 50-58
Open this publication in new window or tab >>Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology
Show others...
2025 (English)In: Experimental and Molecular Medicine, ISSN 1226-3613, E-ISSN 2092-6413, Vol. 57, no 1, p. 50-58Article, review/survey (Refereed) Published
Abstract [en]

Research on pancreatic cancer has transformed with the advent of organoid technology, providing a better platform that closely mimics cancer biology in vivo. This review highlights the critical advancements facilitated by pancreatic organoid models in understanding disease progression, evaluating therapeutic responses, and identifying biomarkers. These three-dimensional cultures enable the proper recapitulation of the cellular architecture and genetic makeup of the original tumors, providing insights into the complex molecular and cellular dynamics at various stages of pancreatic ductal adenocarcinoma (PDAC). We explore the applications of pancreatic organoids in dissecting the tumor microenvironment (TME); elucidating cancer progression, metastasis, and drug resistance mechanisms; and personalizing therapeutic strategies. By overcoming the limitations of traditional 2D cultures and animal models, the use of pancreatic organoids has significantly accelerated translational research, which is promising for improving diagnostic and therapeutic approaches in clinical settings, ultimately aiming to improve the outcomes of patients with pancreatic cancer.

Place, publisher, year, edition, pages
Springer Nature, 2025
National Category
Cancer and Oncology Cell and Molecular Biology
Identifiers
urn:nbn:se:umu:diva-235672 (URN)10.1038/s12276-024-01378-w (DOI)001396135300001 ()39814914 (PubMedID)2-s2.0-85217226319 (Scopus ID)
Available from: 2025-02-24 Created: 2025-02-24 Last updated: 2025-02-24Bibliographically approved
Blomstrand, H., Bodarve, M., Groth, F., Naredi, P., Sund, M., Vilhav, C., . . . Elander, N. O. (2025). Intratumoural expression of dihydropyrimidine dehydrogenase is an independent prognostic factor in resected pancreatic ductal adenocarcinoma treated with adjuvant gemcitabine. Oncology Letters, 29(2), Article ID 99.
Open this publication in new window or tab >>Intratumoural expression of dihydropyrimidine dehydrogenase is an independent prognostic factor in resected pancreatic ductal adenocarcinoma treated with adjuvant gemcitabine
Show others...
2025 (English)In: Oncology Letters, ISSN 1792-1074, E-ISSN 1792-1082, Vol. 29, no 2, article id 99Article in journal (Refereed) Published
Abstract [en]

Pancreatic ductal adenocarcinoma (PDAC) is associated with a poor prognosis, and biomarkers to guide treatment decisions in PDAC are generally lacking. Intratumoural expression of dihydropyrimidine dehydroge- nase (DPD) is a potential prognostic parameter in patients with PDAC undergoing surgical resection and postoperative chemotherapy. In the present study, DPD was analysed by immunohistochemistry of a tissue microarray platform including a real-world cohort of 495 patients with PDAC who had undergone resection with curative intent at any of three tertiary centres, including Northern, Western and Southeastern regions of Sweden, between 1993 and 2019. DPD level (high/low) was analysed and overall survival associations were assessed in treatment subgroups using a multivariate Cox regression model accounting for potential confounders. In patients who had not received adjuvant chemotherapy (n=182), the median overall survival time was 11.6 months (95% CI 9.6-13.5), compared with 28.8 months (25.0-32.6) among those who had (n=313; log-rank P<0.001). The most common type of chemotherapy was gemcitabine single agent (Gem, n=239) followed by gemcitabine plus capecitabine (GemCape, n=39). Tumour-Node-Metastasis (TNM) stage and DPD expression were statistically significant prognostic parameters in the Gem group (HR 1.19, 95% CI 1.01-1.41, P=0.036), with high expres- sion of DPD linked with worse survival. In addition, tumour grade and TNM stage were statistically significant prognostic factors in the group that did not receive any chemotherapy (P≤0.001). No statistically significant parameters were iden- tified in the GemCape group. Taken together, intratumoural expression of DPD may be considered a prognostic marker for patients with PDAC treated with adjuvant gemcitabine following surgical resection, with low expression levels predicting better survival. Further studies in larger cohorts of patients receiving multi-drug or non-gemcitabine based regimens are warranted.

Place, publisher, year, edition, pages
Spandidos Publications, 2025
Keywords
adjuvant chemotherapy, dihydropyrimidine dehydrogenase, gemcitabine, pancreatic cancer, pancreatic ductal carcinoma, prognosis, prognostic factors
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:umu:diva-233391 (URN)10.3892/ol.2024.14845 (DOI)001381167400001 ()39703529 (PubMedID)2-s2.0-85212415469 (Scopus ID)
Funder
Medical Research Council of Southeast Sweden (FORSS), 941207Swedish Society of Medicine, SLS‑960379Region Västerbotten, RV 967602Sjöberg FoundationCancerforskningsfonden i Norrland, LP 23‑2337Cancerforskningsfonden i Norrland, AMP23‑1104Cancerforskningsfonden i Norrland, LP 24‑2377Swedish Cancer Society, 23 2707 PjSwedish Research Council, 2022‑00855Region Västerbotten, RV‑978812Knut and Alice Wallenberg FoundationMarianne and Marcus Wallenberg Foundation, MMW 2020.0189Swedish Research Council, 2019‑01690Swedish Cancer Society, 19 0273Region Västerbotten, RV‑583411Region Västerbotten, RV‑549731Region Västerbotten, RV‑58341Region Västerbotten, RV‑841551Sjöberg Foundation
Available from: 2025-01-09 Created: 2025-01-09 Last updated: 2025-03-21Bibliographically approved
Löhr, J.-M., Öhlund, D., Söreskog, E., Andersson, E., Vujasinovic, M., Zethraeus, N. & Sund, M. (2024). Can our experience with surveillance for inherited pancreatic cancer help to identify early pancreatic cancer in the general population?. Familial Cancer, 23(3), 399-403
Open this publication in new window or tab >>Can our experience with surveillance for inherited pancreatic cancer help to identify early pancreatic cancer in the general population?
Show others...
2024 (English)In: Familial Cancer, ISSN 1389-9600, E-ISSN 1573-7292, Vol. 23, no 3, p. 399-403Article, review/survey (Refereed) Published
Abstract [en]

Screening of the general population for cancer is a matter of primary prevention reducing the burden of disease. Whilst this is successful for several cancers including breast, colon and prostate, the situation to screen and hence prevent pancreatic cancer is different. The organ is not as accessible to simple physical exam or biological samples (fecal or blood test). Neither exists a blood test such as PSA that is cost-effective. Reviewing the evidence from screening risk groups for pancreatic cancer, one must conclude that there is no rational at present to screen the general population, for a lack of appropriate tests.

Place, publisher, year, edition, pages
Springer Nature, 2024
Keywords
General population, Incidental finding, Individuals at risk, Pancreatic cancer, Screening, Surveillance
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:umu:diva-222299 (URN)10.1007/s10689-024-00363-6 (DOI)001174923700001 ()38441833 (PubMedID)2-s2.0-85186597174 (Scopus ID)
Funder
Signe and Olof Wallenius FoundationSwedish Cancer Society, 21 1585Swedish Cancer Society, 22 2318Swedish Cancer Society, 19 0273Swedish Cancer Society, 2011/1110Swedish Cancer Society, 2016/634Swedish Cancer Society, 2017/332Swedish Cancer Society, 2017/827Swedish Cancer Society, 20 1339Swedish Research Council, 2016–02990Swedish Research Council, 2019−01690Region Västerbotten, 583411Region Västerbotten, RV-583411Region Västerbotten, RV-549731Region Västerbotten, RV-841551Region Västerbotten, RV-930478Region Västerbotten, RV-930132Region Västerbotten, RV-99607108Region Västerbotten, RV-9960708Region Västerbotten, VLL-837731Region Västerbotten, RV-930167Region Västerbotten, VLL- 643451Region Västerbotten, RV-978812Sjöberg FoundationKnut and Alice Wallenberg FoundationMarianne and Marcus Wallenberg Foundation
Note

Special issue

Available from: 2024-03-14 Created: 2024-03-14 Last updated: 2024-08-20Bibliographically approved
Vlăduț, C., Steiner, C., Löhr, M., Gökçe, D. T., Maisonneuve, P., Hank, T., . . . Hoogenboom, S. A. (2024). High prevalence of pancreatic steatosis in pancreatic cancer patients: a meta-analysis and systematic review. Pancreatology (Print)
Open this publication in new window or tab >>High prevalence of pancreatic steatosis in pancreatic cancer patients: a meta-analysis and systematic review
Show others...
2024 (English)In: Pancreatology (Print), ISSN 1424-3903, E-ISSN 1424-3911Article in journal (Refereed) Epub ahead of print
Abstract [en]

Objective: In the last decade there has been increasing interest in defining pancreatic steatosis (PS) and establishing its association with pancreatic ductal adenocarcinoma (PDAC). However, no consensus guidelines have yet been published on the management of PS. In this systematic review and meta-analysis performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we investigated the association between PS and PDAC.

Design: Medical literature between 2007 and 2023 was reviewed for eligible trials investigating the prevalence of PS in patients with PDAC. Eligible studies reporting on PS, assessed via imaging or histology, were included. The primary objective was to determine the association between PDAC and PS by comparing the prevalence of PS in individuals with- and without PDAC. Secondary, an evaluation was conducted to establish whether the method of assessment correlated with the association of PDAC and PS, and the prevalence of PDAC in individuals with PS. Measures of effect size were determined using odds ratios (ORs) and corresponding 95 % confidence intervals (95 % CI).

Results: The systematic review identified a total of 23 studies, of which seventeen studies examined PS prevalence among PDAC patients and were included in the meta-analysis. Overall, the pooled prevalence of PS in patients with PDAC was 53.6 % (95 % CI 40.9–66.2). No significant difference in PS prevalence was observed across various diagnostic methods or geographical regions. Overall, the pooled OR for PS in patients with PDAC compared to controls was 3.23 (95 % CI 1.86–5.60).

Conclusions: PDAC patients have a high prevalence of PS, and they are significantly more likely to have PS compared to controls. These findings emphasize the need to prioritize a standardized approach to the diagnosis, follow-up, and treatment of PS, with future studies focusing on identifying patients who would benefit from PDAC surveillance programs.

Place, publisher, year, edition, pages
Elsevier, 2024
Keywords
Fatty pancreas, NAFPD, Obesity, Pancreatic ductal adenocarcinoma, Pancreatic steatosis
National Category
Gastroenterology and Hepatology
Identifiers
urn:nbn:se:umu:diva-233398 (URN)10.1016/j.pan.2024.11.010 (DOI)001423084300001 ()2-s2.0-85212661619 (Scopus ID)
Available from: 2025-01-08 Created: 2025-01-08 Last updated: 2025-04-24
Borgmästars, E., Jacobson, S., Simm, M., Johansson, M., Billing, O., Lundin, C., . . . Sund, M. (2024). Metabolomics for early pancreatic cancer detection in plasma samples from a Swedish prospective population-based biobank. Journal of Gastrointestinal Oncology, 15(2), 755-767
Open this publication in new window or tab >>Metabolomics for early pancreatic cancer detection in plasma samples from a Swedish prospective population-based biobank
Show others...
2024 (English)In: Journal of Gastrointestinal Oncology, ISSN 2078-6891, E-ISSN 2219-679X, Vol. 15, no 2, p. 755-767Article in journal (Refereed) Published
Abstract [en]

Background: Pancreatic ductal adenocarcinoma (pancreatic cancer) is often detected at late stages resulting in poor overall survival. To improve survival, more patients need to be diagnosed early when curative surgery is feasible. We aimed to identify circulating metabolites that could be used as early pancreatic cancer biomarkers.

Methods: We performed metabolomics by liquid and gas chromatography-mass spectrometry in plasma samples from 82 future pancreatic cancer patients and 82 matched healthy controls within the Northern Sweden Health and Disease Study (NSHDS). Logistic regression was used to assess univariate associations between metabolites and pancreatic cancer risk. Least absolute shrinkage and selection operator (LASSO) logistic regression was used to design a metabolite-based risk score. We used receiver operating characteristic (ROC) analyses to assess the discriminative performance of the metabolite-based risk score.

Results: Among twelve risk-associated metabolites with a nominal P value <0.05, we defined a risk score of three metabolites [indoleacetate, 3-hydroxydecanoate (10:0-OH), and retention index (RI): 2,745.4] using LASSO. A logistic regression model containing these three metabolites, age, sex, body mass index (BMI), smoking status, sample date, fasting status, and carbohydrate antigen 19-9 (CA 19-9) yielded an internal area under curve (AUC) of 0.784 [95% confidence interval (CI): 0.714–0.854] compared to 0.681 (95% CI: 0.597–0.764) for a model without these metabolites (P value =0.007). Seventeen metabolites were significantly associated with pancreatic cancer survival [false discovery rate (FDR) <0.1].

Conclusions: Indoleacetate, 3-hydroxydecanoate (10:0-OH), and RI: 2,745.4 were identified as the top candidate biomarkers for early detection. However, continued efforts are warranted to determine the usefulness of these metabolites as early pancreatic cancer biomarkers.

Place, publisher, year, edition, pages
AME Publishing Company, 2024
Keywords
biomarkers, hyperglycemia, Pancreatic neoplasms, risk, survival
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:umu:diva-224962 (URN)10.21037/jgo-23-930 (DOI)001284655300018 ()2-s2.0-85192826642 (Scopus ID)
Funder
Umeå UniversitySwedish Cancer Society, 19 0273Swedish Cancer Society, 2017-557Swedish Cancer Society, CAN 2017/332Swedish Cancer Society, CAN 2017/827Swedish Research Council, 2019-01690Swedish Research Council, 2016-02990Swedish Research Council, 2017-01531Region Västerbotten, RV-583411Region Västerbotten, RV-549731Region Västerbotten, RV-841551Region Västerbotten, RV-930167Region Västerbotten, VLL-643451
Available from: 2024-05-27 Created: 2024-05-27 Last updated: 2025-04-24Bibliographically approved
Labori, K. J., Bratlie, S. O., Andersson, B., Angelsen, J.-H., Biörserud, C., Björnsson, B., . . . Halimi, A. (2024). Neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer (NORPACT-1): a multicentre, randomised, phase 2 trial. The Lancet Gastroenterology & Hepatology, 9(3), 205-217
Open this publication in new window or tab >>Neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer (NORPACT-1): a multicentre, randomised, phase 2 trial
Show others...
2024 (English)In: The Lancet Gastroenterology & Hepatology, ISSN 2468-1253, Vol. 9, no 3, p. 205-217Article in journal (Refereed) Published
Abstract [en]

Background: In patients undergoing resection for pancreatic cancer, adjuvant modified fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) improves overall survival compared with alternative chemotherapy regimens. We aimed to compare the efficacy and safety of neoadjuvant FOLFIRINOX with the standard strategy of upfront surgery in patients with resectable pancreatic ductal adenocarcinoma.

Methods: NORPACT-1 was a multicentre, randomised, phase 2 trial done in 12 hospitals in Denmark, Finland, Norway, and Sweden. Eligible patients were aged 18 years or older, with a WHO performance status of 0 or 1, and had a resectable tumour of the pancreatic head radiologically strongly suspected to be pancreatic adenocarcinoma. Participants were randomly assigned (3:2 before October, 2018, and 1:1 after) to the neoadjuvant FOLFIRINOX group or upfront surgery group. Patients in the neoadjuvant FOLFIRINOX group received four neoadjuvant cycles of FOLFIRINOX (oxaliplatin 85 mg/m2, irinotecan 180 mg/m2, leucovorin 400 mg/m2, and fluorouracil 400 mg/m2 bolus then 2400 mg/m2 over 46 h on day 1 of each 14-day cycle), followed by surgery and adjuvant chemotherapy. Patients in the upfront surgery group underwent surgery and then received adjuvant chemotherapy. Initially, adjuvant chemotherapy was gemcitabine plus capecitabine (gemcitabine 1000 mg/m2 over 30 min on days 1, 8, and 15 of each 28-day cycle and capecitabine 830 mg/m2 twice daily for 3 weeks with 1 week of rest in each 28-day cycle; four cycles in the neoadjuvant FOLFIRINOX group, six cycles in the upfront surgery group). A protocol amendment was subsequently made to permit use of adjuvant modified FOLFIRINOX (oxaliplatin 85 mg/m2, irinotecan 150 mg/m2, leucovorin 400 mg/m2, and fluorouracil 2400 mg/m2 over 46 h on day 1 of each 14-day cycle; eight cycles in the neoadjuvant FOLFIRINOX group, 12 cycles in the upfront surgery group). Randomisation was performed with a computerised algorithm that stratified for each participating centre and used a concealed block size of two to six. Patients, investigators, and study team members were not masked to treatment allocation. The primary endpoint was overall survival at 18 months. Analyses were done in the intention-to-treat (ITT) and per-protocol populations. Safety was assessed in all patients who were randomly assigned and received at least one cycle of neoadjuvant or adjuvant therapy. This trial is registered with ClinicalTrials.gov, NCT02919787, and EudraCT, 2015-001635-21, and is ongoing.

Findings: Between Feb 8, 2017, and April 21, 2021, 77 patients were randomly assigned to receive neoadjuvant FOLFIRINOX and 63 to undergo upfront surgery. All patients were included in the ITT analysis. For the per-protocol analysis, 17 (22%) patients were excluded from the neoadjuvant FOLFIRINOX group (ten did not receive neoadjuvant therapy, four did not have pancreatic ductal adenocarcinoma, and three received another neoadjuvant regimen), and eight (13%) were excluded from the upfront surgery group (seven did not have pancreatic ductal adenocarcinoma and one did not undergo surgical exploration). 61 (79%) of 77 patients in the neoadjuvant FOLFIRINOX group received neoadjuvant therapy. The proportion of patients alive at 18 months by ITT was 60% (95% CI 49–71) in the neoadjuvant FOLFIRINOX group versus 73% (62–84) in the upfront surgery group (p=0·032), and median overall survival by ITT was 25·1 months (95% CI 17·2–34·9) versus 38·5 months (27·6–not reached; hazard ratio [HR] 1·52 [95% CI 1·00–2·33], log-rank p=0·050). The proportion of patients alive at 18 months in per-protocol analysis was 57% (95% CI 46–67) in the neoadjuvant FOLFIRINOX group versus 70% (55–83) in the upfront surgery group (p=0·14), and median overall survival in per-protocol population was 23·0 months (95% CI 16·2–34·9) versus 34·4 months (19·4–not reached; HR 1·46 [95% CI 0·99–2·17], log-rank p=0·058). In the safety population, 42 (58%) of 73 patients in the neoadjuvant FOLFIRINOX group and 19 (40%) of 47 patients in the upfront surgery group had at least one grade 3 or worse adverse event. 63 (82%) of 77 patients in the neoadjuvant group and 56 (89%) of 63 patients in the upfront surgery group had resection (p=0·24). One sudden death of unknown cause and one COVID-19-related death occurred after the first cycle of neoadjuvant FOLFIRINOX. Adjuvant chemotherapy was initiated in 51 (86%) of 59 patients with resected pancreatic ductal adenocarcinoma in the neoadjuvant FOLFIRINOX group and 44 (90%) of 49 patients with resected pancreatic ductal adenocarcinoma in the upfront surgery group (p=0·56). Adjuvant modified FOLFIRINOX was given to 13 (25%) patients in the neoadjuvant FOLFIRINOX group and 19 (43%) patients in the upfront surgery group. During adjuvant chemotherapy, neutropenia (11 [22%] patients in the neoadjuvant FOLFIRINOX group and five [11%] in the upfront surgery group) was the most common grade 3 or worse adverse event.

Interpretation: This phase 2 trial did not show a survival benefit from neoadjuvant FOLFIRINOX in resectable pancreatic ductal adenocarcinoma compared with upfront surgery. Implementation of neoadjuvant FOLFIRINOX was challenging. Future trials on treatment sequencing in resectable pancreatic ductal adenocarcinoma should be biomarker driven.

Place, publisher, year, edition, pages
Elsevier, 2024
National Category
Cancer and Oncology Surgery
Identifiers
urn:nbn:se:umu:diva-221022 (URN)10.1016/S2468-1253(23)00405-3 (DOI)001184600000001 ()38237621 (PubMedID)2-s2.0-85183761228 (Scopus ID)
Funder
Sjöberg Foundation
Available from: 2024-03-06 Created: 2024-03-06 Last updated: 2025-04-24Bibliographically approved
Espona-Fiedler, M., Patthey, C., Lindblad, S., Sarró, I. & Öhlund, D. (2024). Overcoming therapy resistance in pancreatic cancer: new insights and future directions. Biochemical Pharmacology, 229, Article ID 116492.
Open this publication in new window or tab >>Overcoming therapy resistance in pancreatic cancer: new insights and future directions
Show others...
2024 (English)In: Biochemical Pharmacology, ISSN 0006-2952, E-ISSN 1356-1839, Vol. 229, article id 116492Article, review/survey (Refereed) Published
Abstract [en]

Pancreatic adenocarcinoma (PDAC) is predicted to become the second leading cause of cancer deaths by 2030 and this is mostly due to therapy failure. Limited treatment options and resistance to standard-of-care (SoC) therapies makes PDAC one of the cancer types with poorest prognosis and survival rates [1,2]. Pancreatic tumors are renowned for their poor response to therapeutic interventions including targeted therapies, chemotherapy and radiotherapy. Herein, we review hallmarks of therapy resistance in PDAC and current strategies aiming to tackle escape mechanisms and to re-sensitize cancer cells to therapy. We will further provide insights on recent advances in the field of drug discovery, nanomedicine, and disease models that are setting the ground for future research.

Place, publisher, year, edition, pages
Elsevier, 2024
Keywords
Cancer-associated fibroblast, Pancreatic cancer, Therapy resistance, Tumor microenvironment
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:umu:diva-228901 (URN)10.1016/j.bcp.2024.116492 (DOI)001298078900001 ()39153553 (PubMedID)2-s2.0-85201605382 (Scopus ID)
Funder
Cancerforskningsfonden i Norrland, AMP23-1104Swedish Cancer Society, 23 2707 PjCancerforskningsfonden i Norrland, LP 24-2377Swedish Research Council, 2022-00855Region Västerbotten, RV-978812Knut and Alice Wallenberg FoundationMarianne and Marcus Wallenberg Foundation, MMW 2020.0189
Available from: 2024-09-02 Created: 2024-09-02 Last updated: 2025-04-24Bibliographically approved
Bermejo-Rodriguez, C., Araos Henríquez, J., Caligiuri, G., Pinto Teles, S., Park, Y., Evans, A., . . . Perez-Mancera, P. A. (2024). Scribble deficiency promotes pancreatic ductal adenocarcinoma development and metastasis. Cancer Research, 84(18), 2968-2984
Open this publication in new window or tab >>Scribble deficiency promotes pancreatic ductal adenocarcinoma development and metastasis
Show others...
2024 (English)In: Cancer Research, ISSN 0008-5472, E-ISSN 1538-7445, Vol. 84, no 18, p. 2968-2984Article in journal (Refereed) Published
Abstract [en]

Perturbation of cell polarity is a hallmark of pancreatic ductal adenocarcinoma (PDAC) progression. Scribble (SCRIB) is a well-characterized polarity regulator that has diverse roles in the pathogenesis of human neoplasms. To investigate the impact of SCRIB deficiency in PDAC development and progression, Scrib expression was genetically ablated in well-established mouse models of PDAC. Scrib loss in combination with KrasG12D did not influence development of pancreatic intraepithelial neoplasms in mice. However, Scrib deletion cooperated with KrasG12D and concomitant Trp53 heterozygous deletion to promote invasive PDAC and metastatic dissemination, leading to reduced overall survival. Immunohistochemical and transcriptome analyses revealed that Scrib-null tumors display a pronounced reduction of collagen content and an abundance of cancer-associated fibroblasts (CAF). Mechanistically, IL1α levels were reduced in Scrib-deficient tumors, and Scrib knockdown downregulated IL1α in mouse PDAC organoids (mPDO), which impaired CAF activation. Furthermore, Scrib loss increased YAP activation in mPDOs and established PDAC cell lines, enhancing cell survival. Clinically, SCRIB expression was decreased in human PDAC, and SCRIB mislocalization was associated with poorer patient outcome. These results indicate that SCRIB deficiency enhances cancer cell survival and remodels the tumor microenvironment to accelerate PDAC development and progression, establishing the tumor suppressor function of SCRIB in advanced pancreatic cancer.  

Significance: SCRIB loss promotes invasive pancreatic cancer development via both cell-autonomous and non-cell-autonomous processes and is associated with poorer outcomes, denoting SCRIB as a tumor suppressor and potential biomarker for the prediction of recurrence.

Place, publisher, year, edition, pages
American Association For Cancer Research (AACR), 2024
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:umu:diva-230017 (URN)10.1158/0008-5472.CAN-23-3419 (DOI)001313818100005 ()2-s2.0-85204258425 (Scopus ID)
Funder
NIH (National Institutes of Health), 5P30CA45508NIH (National Institutes of Health), R01CA249002NIH (National Institutes of Health), U01CA224013NIH (National Institutes of Health), U01CA210240NIH (National Institutes of Health), 1R01CA188134
Available from: 2024-10-04 Created: 2024-10-04 Last updated: 2024-10-08Bibliographically approved
Deiana, M., Andrés Castán, J. M., Josse, P., Kahsay, A., Sánchez, D. P., Morice, K., . . . Sabouri, N. (2023). A new G-quadruplex-specific photosensitizer inducing genome instability in cancer cells by triggering oxidative DNA damage and impeding replication fork progression. Nucleic Acids Research, 51(12), 6264-6285
Open this publication in new window or tab >>A new G-quadruplex-specific photosensitizer inducing genome instability in cancer cells by triggering oxidative DNA damage and impeding replication fork progression
Show others...
2023 (English)In: Nucleic Acids Research, ISSN 0305-1048, E-ISSN 1362-4962, Vol. 51, no 12, p. 6264-6285Article in journal (Refereed) Published
Abstract [en]

Photodynamic therapy (PDT) ideally relies on the administration, selective accumulation and photoactivation of a photosensitizer (PS) into diseased tissues. In this context, we report a new heavy-atom-free fluorescent G-quadruplex (G4) DNA-binding PS, named DBI. We reveal by fluorescence microscopy that DBI preferentially localizes in intraluminal vesicles (ILVs), precursors of exosomes, which are key components of cancer cell proliferation. Moreover, purified exosomal DNA was recognized by a G4-specific antibody, thus highlighting the presence of such G4-forming sequences in the vesicles. Despite the absence of fluorescence signal from DBI in nuclei, light-irradiated DBI-treated cells generated reactive oxygen species (ROS), triggering a 3-fold increase of nuclear G4 foci, slowing fork progression and elevated levels of both DNA base damage, 8-oxoguanine, and double-stranded DNA breaks. Consequently, DBI was found to exert significant phototoxic effects (at nanomolar scale) toward cancer cell lines and tumor organoids. Furthermore, in vivo testing reveals that photoactivation of DBI induces not only G4 formation and DNA damage but also apoptosis in zebrafish, specifically in the area where DBI had accumulated. Collectively, this approach shows significant promise for image-guided PDT.

Place, publisher, year, edition, pages
Oxford University Press, 2023
National Category
Biochemistry Molecular Biology Cell and Molecular Biology
Identifiers
urn:nbn:se:umu:diva-212227 (URN)10.1093/nar/gkad365 (DOI)000988008500001 ()37191066 (PubMedID)2-s2.0-85164253573 (Scopus ID)
Funder
Swedish Cancer Society, 22 2380 PjSwedish Research Council, VR-MH 2021–02468Knut and Alice Wallenberg Foundation, KAW 2021-0173Swedish Cancer Society, 21 0302 PT 01 HWenner-Gren Foundations, UPD2020-0097Swedish Cancer Society, 20 0827 PjFCancerforskningsfonden i Norrland, LP 22-2312Cancerforskningsfonden i Norrland, LP20 1024 2257Cancerforskningsfonden i Norrland, LP 21–2298Swedish Research Council, 2017-01531Swedish Society of Medicine, SLS-890521Region Västerbotten, RV-930167Sjöberg FoundationKnut and Alice Wallenberg Foundation, KAW 2015.0114Marianne and Marcus Wallenberg Foundation, MMW 2020.0189Swedish Cancer Society, 20 1339 PjF
Available from: 2023-07-21 Created: 2023-07-21 Last updated: 2025-02-20Bibliographically approved
Gonzalez-Castrillon, L. M., Wurmser, M., Öhlund, D. & Wilson, S. I. (2023). Dysregulation of core neurodevelopmental pathways: a common feature of cancers with perineural invasion. Frontiers in Genetics, 14, Article ID 1181775.
Open this publication in new window or tab >>Dysregulation of core neurodevelopmental pathways: a common feature of cancers with perineural invasion
2023 (English)In: Frontiers in Genetics, E-ISSN 1664-8021, Vol. 14, article id 1181775Article in journal (Refereed) Published
Abstract [en]

Background: High nerve density in tumors and metastasis via nerves (perineural invasion—PNI) have been reported extensively in solid tumors throughout the body including pancreatic, head and neck, gastric, prostate, breast, and colorectal cancers. Ablation of tumor nerves results in improved disease outcomes, suggesting that blocking nerve–tumor communication could be a novel treatment strategy. However, the molecular mechanisms underlying this remain poorly understood. Thus, the aim here was to identify molecular pathways underlying nerve–tumor crosstalk and to determine common molecular features between PNI-associated cancers.

Results: Analysis of head and neck (HNSCC), pancreatic, and gastric (STAD) cancer Gene Expression Omnibus datasets was used to identify differentially expressed genes (DEGs). This revealed extracellular matrix components as highly dysregulated. To enrich for pathways associated with PNI, genes previously correlated with PNI in STAD and in 2 HNSCC studies where tumor samples were segregated by PNI status were analyzed. Neurodevelopmental genes were found to be enriched with PNI. In datasets where tumor samples were not segregated by PNI, neurodevelopmental pathways accounted for 12%–16% of the DEGs. Further dysregulation of axon guidance genes was common to all cancers analyzed. By examining paralog genes, a clear pattern emerged where at least one family member from several axon guidance pathways was affected in all cancers examined. Overall 17 different axon guidance gene families were disrupted, including the ephrin–Eph, semaphorin–neuropilin/plexin, and slit–robo pathways. These findings were validated using The Cancer Genome Atlas and cross-referenced to other cancers with a high incidence of PNI including colon, cholangiocarcinoma, prostate, and breast cancers. Survival analysis revealed that the expression levels of neurodevelopmental gene families impacted disease survival.

Conclusion: These data highlight the importance of the tumor as a source of signals for neural tropism and neural plasticity as a common feature of cancer. The analysis supports the hypothesis that dysregulation of neurodevelopmental programs is a common feature associated with PNI. Furthermore, the data suggested that different cancers may have evolved to employ alternative genetic strategies to disrupt the same pathways. Overall, these findings provide potential druggable targets for novel therapies of cancer management and provide multi-cancer molecular biomarkers.

Place, publisher, year, edition, pages
Frontiers Media S.A., 2023
Keywords
bioinformatics, biomarker, cancer, head and neck squamous cell carcinoma, neurodevelopment, pancreatic ductal adenocarcinoma, perineural invasion, stomach adenocarcinoma
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:umu:diva-214621 (URN)10.3389/fgene.2023.1181775 (DOI)001065686900001 ()2-s2.0-85170839267 (Scopus ID)
Funder
Umeå University, FS 2.1.6-1119-19Knut and Alice Wallenberg Foundation, MMW 2020.0189Marianne and Marcus Wallenberg Foundation, MMW 2020.0189
Available from: 2023-09-26 Created: 2023-09-26 Last updated: 2025-04-24Bibliographically approved
Projects
Targeting Tumor-stromal Interactions in Pancreatic Cancer [2017-01531_VR]; Umeå University; Publications
Lidström, T., Cumming, J., Gaur, R., Frängsmyr, L., Pateras, I., Mickert, M. J., . . . Öhlund, D. (2023). Extracellular galectin 4 drives immune evasion and promotes T-cell apoptosis in pancreatic cancer. Cancer immunology research, 11(1), 72-92Lidström, T. (2022). Immunosuppressive and metastasis-promoting matrisome proteins in pancreatic cancer: the role of galectin 4 and SERPINB5. (Doctoral dissertation). Umeå: Umeå UniversityLidström, T., Patthey, C. & Öhlund, D.Coordination and cooperation of immunosuppressive mechanisms in pancreatic ductal adenocarcinoma.
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-5847-2778

Search in DiVA

Show all publications