Logo: to the web site of Uppsala University

uu.sePublications from Uppsala University
System disruptions
We are currently experiencing disruptions on the search portals due to high traffic. We are working to resolve the issue, you may temporarily encounter an error message.
Change search
Link to record
Permanent link

Direct link
Alternative names
Publications (10 of 85) Show all publications
Spiegelberg, D., Hwang, L.-A., Pua, K. H., Kumar, S. C., Koh, X. Y., Koh, X. H., . . . Lane, D. (2025). Targeting mutant p53: Evaluation of novel anti-p53R175H monoclonal antibodies as diagnostic tools. Scientific Reports, 15(1), Article ID 1000.
Open this publication in new window or tab >>Targeting mutant p53: Evaluation of novel anti-p53R175H monoclonal antibodies as diagnostic tools
Show others...
2025 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 15, no 1, article id 1000Article in journal (Refereed) Published
Abstract [en]

About 50% of all cancers carry a mutation in p53 that impairs its tumor suppressor function. The p53 missense mutation p53R175H (p53R172H in mice) is a hotspot mutation in various cancer types. Therefore, monoclonal antibodies selectively targeting clinically relevant mutations like p53R175H could prove immensely value. We aimed to evaluate the in vitro and in vivo binding properties of two novel anti-p53R175H monoclonal antibodies and to assess their performance as agents for molecular imaging. In vitro, 125I-4H5 and 125I-7B9 demonstrated long shelf life and antigen-specific binding. Our in vivo study design allowed head-to-head comparison of the antibodies in a double tumor model using repeated SPECT/CT imaging, followed by biodistribution and autoradiography. Both tracers performed similarly, with marginally faster blood clearance for 125I-7B9. Repeated molecular imaging demonstrated suitable imaging characteristics for both antibodies, with the best contrast images occurring at 48 h post-injection. Significantly higher uptake was detected in the mut-p53-expressing tumors, confirmed by ex vivo autoradiography. We conclude that molecular imaging with an anti-p53R175H tracer could be a promising approach for cancer diagnostics and could be further applied for patient stratification and treatment response monitoring of mutant p53-targeted therapeutics.

Place, publisher, year, edition, pages
Springer Nature, 2025
Keywords
Cancer diagnostics, Mutant p53, Intracellular targets, Molecular imaging
National Category
Cancer and Oncology Radiology, Nuclear Medicine and Medical Imaging Hematology
Identifiers
urn:nbn:se:uu:diva-549506 (URN)10.1038/s41598-024-83871-w (DOI)001398315800007 ()39762369 (PubMedID)2-s2.0-85214268703 (Scopus ID)
Funder
Swedish Cancer Society, 21 0371 FESwedish Cancer Society, 24 3787 PjSwedish Cancer Society, 24 3485 PjSwedish Childhood Cancer Foundation, FT2023-0023Swedish Childhood Cancer Foundation, PR2023-0111Swedish Research Council, 2024-03447
Available from: 2025-02-20 Created: 2025-02-20 Last updated: 2025-02-20Bibliographically approved
Chandramohan, A., Josien, H., Yuen, T. Y., Duggal, R., Spiegelberg, D., Yan, L., . . . Partridge, A. W. (2024). Design-Rules for Stapled Alpha-Helical Peptides with On-Target In Vivo Activity: Application to Mdm2/X dual antagonists. Nature Communications, 15(1), Article ID 489.
Open this publication in new window or tab >>Design-Rules for Stapled Alpha-Helical Peptides with On-Target In Vivo Activity: Application to Mdm2/X dual antagonists
Show others...
2024 (English)In: Nature Communications, E-ISSN 2041-1723, Vol. 15, no 1, article id 489Article in journal (Refereed) Published
Abstract [en]

Although stapled α-helical peptides can address challenging targets, their advancement is impeded by poor understandings for making them cell permeable while avoiding off-target toxicities. By synthesizing >350 molecules, we present workflows for identifying stapled peptides against Mdm2(X) with in vivo activity and no off-target effects. Key insights include a clear correlation between lipophilicity and permeability, removal of positive charge to avoid off-target toxicities, judicious anionic residue placement to enhance solubility/behavior, optimization of C-terminal length/helicity to enhance potency, and optimization of staple type/number to avoid polypharmacology. Workflow application gives peptides with >292x improved cell proliferation potencies and no off-target cell proliferation effects ( > 3800x on-target index). Application of these ‘design rules’ to a distinct Mdm2(X) peptide series improves ( > 150x) cellular potencies and removes off-target toxicities. The outlined workflow should facilitate therapeutic impacts, especially for those targets such as Mdm2(X) that have hydrophobic interfaces and are targetable with a helical motif.

Place, publisher, year, edition, pages
Springer Nature, 2024
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:uu:diva-515080 (URN)10.1038/s41467-023-43346-4 (DOI)001142873900002 ()38216578 (PubMedID)
Available from: 2023-10-26 Created: 2023-10-26 Last updated: 2024-10-25Bibliographically approved
Chandramohan, A., Josien, H., Yuen, T. Y., Duggal, R., Spiegelberg, D., Yan, L., . . . Partridge, A. W. (2024). Design-rules for stapled peptides with in vivo activity and their application to Mdm2/X antagonists. Nature Communications, 15(1), Article ID 489.
Open this publication in new window or tab >>Design-rules for stapled peptides with in vivo activity and their application to Mdm2/X antagonists
Show others...
2024 (English)In: Nature Communications, E-ISSN 2041-1723, Vol. 15, no 1, article id 489Article in journal (Refereed) Published
Abstract [en]

Although stapled α-helical peptides can address challenging targets, their advancement is impeded by poor understandings for making them cell permeable while avoiding off-target toxicities. By synthesizing >350 molecules, we present workflows for identifying stapled peptides against Mdm2(X) with in vivo activity and no off-target effects. Key insights include a clear correlation between lipophilicity and permeability, removal of positive charge to avoid off-target toxicities, judicious anionic residue placement to enhance solubility/behavior, optimization of C-terminal length/helicity to enhance potency, and optimization of staple type/number to avoid polypharmacology. Workflow application gives peptides with >292x improved cell proliferation potencies and no off-target cell proliferation effects ( > 3800x on-target index). Application of these ‘design rules’ to a distinct Mdm2(X) peptide series improves ( > 150x) cellular potencies and removes off-target toxicities. The outlined workflow should facilitate therapeutic impacts, especially for those targets such as Mdm2(X) that have hydrophobic interfaces and are targetable with a helical motif.

Place, publisher, year, edition, pages
Springer Nature, 2024
National Category
Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:uu:diva-521843 (URN)10.1038/s41467-023-43346-4 (DOI)001142873900002 ()38216578 (PubMedID)
Available from: 2024-01-29 Created: 2024-01-29 Last updated: 2024-01-29Bibliographically approved
Ingelshed, K., Melssen, M. M., Kannan, P., Chandramohan, A., Partridge, A. W., Jiang, L., . . . Spiegelberg, D. (2024). MDM2/MDMX inhibition by Sulanemadlin synergizes with anti-Programmed Death 1 immunotherapy in wild-type p53 tumors. iScience, 27(6), Article ID 109862.
Open this publication in new window or tab >>MDM2/MDMX inhibition by Sulanemadlin synergizes with anti-Programmed Death 1 immunotherapy in wild-type p53 tumors
Show others...
2024 (English)In: iScience, E-ISSN 2589-0042, Vol. 27, no 6, article id 109862Article in journal (Refereed) Published
Abstract [en]

Immunotherapy has revolutionized cancer treatment but its efficacy depends on a robust immune response in the tumor. Silencing of the tumor suppressor p53 is common in tumors and can affect the recruitment and activation of different immune cells, leading to immune evasion and poor therapy response. We found that the p53 activating stapled peptide MDM2/MDMX inhibitor Sulanemadlin (ALRN-6924) inhibited p53 wild-type cancer cell growth in vitro and in vivo. In mice carrying p53 wild-type CT26.WT tumors, monotherapy with the PD-1 inhibitor DX400 or Sulanemadlin delayed tumor doubling time by 50% and 37%, respectively, while combination therapy decreased tumor doubling time by 93% leading to an increased median survival time. Sulanemadlin treatment led to increased immunogenicity and combination treatment with PD-1 inhibition resulted in an increased tumor infiltration of lymphocytes. This combination treatment strategy could potentially turn partial responders into responders of immunotherapy, expanding the patient target group for PD-1-targeting immunotherapy.

Place, publisher, year, edition, pages
Elsevier, 2024
Keywords
microenvironment, Molecular biology, Immunology, cancer
National Category
Cancer and Oncology Immunology in the medical area
Identifiers
urn:nbn:se:uu:diva-533024 (URN)10.1016/j.isci.2024.109862 (DOI)001243716300001 ()38784022 (PubMedID)
Funder
Karolinska InstituteSwedish Cancer Society, 21 0371 FESwedish Research Council, 2013-08807Swedish Research Council, 2021-03178Swedish Research Council, 2020-01377Swedish Cancer Society, 20 1114 PjFSwedish Cancer Society, 22 0546 SIASwedish Cancer Society, CAN 21/1534Swedish Cancer Society, CAN 200191Swedish Childhood Cancer Foundation, FT2023-0023Swedish Childhood Cancer Foundation, PR2023-0111Erik, Karin och Gösta Selanders FoundationÅke Wiberg FoundationSwedish Childhood Cancer Foundation, TJ2022-0016Swedish Childhood Cancer Foundation, PR2023-0033
Available from: 2024-06-25 Created: 2024-06-25 Last updated: 2024-11-18Bibliographically approved
Sarén, T., Saronio, G., Marti Torrell, P., Zhu, X., Thelander, J., Andersson, Y., . . . Essand, M. (2023). Complementarity-determining region clustering may cause CAR-T cell dysfunction. Nature Communications, 14, Article ID 4732.
Open this publication in new window or tab >>Complementarity-determining region clustering may cause CAR-T cell dysfunction
Show others...
2023 (English)In: Nature Communications, E-ISSN 2041-1723, Vol. 14, article id 4732Article in journal (Refereed) Published
Abstract [en]

Chimeric antigen receptor (CAR)-T cell therapy is rapidly advancing as cancer treatment, however, designing an optimal CAR remains challenging. A single-chain variable fragment (scFv) is generally used as CAR targeting moiety, wherein the complementarity-determining regions (CDRs) define its specificity. We report here that the CDR loops can cause CAR clustering, leading to antigen-independent tonic signalling and subsequent CAR-T cell dysfunction. We show via CARs incorporating scFvs with identical framework and varying CDR sequences that CARs may cluster on the T cell surface, which leads to antigen-independent CAR-T cell activation, characterized by increased cell size and interferon (IFN)-& gamma; secretion. This results in CAR-T cell exhaustion, activation-induced cell death and reduced responsiveness to target-antigen-expressing tumour cells. CDR mutagenesis confirms that the CAR-clustering is mediated by CDR-loops. In summary, antigen-independent tonic signalling can be induced by CDR-mediated CAR clustering, which could not be predicted from the scFv sequences, but could be tested for by evaluating the activity of unstimulated CAR-T cells.

Place, publisher, year, edition, pages
Springer Nature, 2023
National Category
Immunology in the medical area Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-510646 (URN)10.1038/s41467-023-40303-z (DOI)001050351300037 ()37563127 (PubMedID)
Funder
Swedish Research Council, 2019-01326Swedish Cancer Society, 19 0184PjSwedish Childhood Cancer Foundation, TJ 2019-0014Swedish Childhood Cancer Foundation, PR2020-0167Sjöberg Foundation, 2020-01-07-06Science for Life Laboratory, SciLifeLab, DP_ME_107DP_ME_107
Available from: 2023-09-04 Created: 2023-09-04 Last updated: 2023-09-04Bibliographically approved
Mohajershojai, T., Spangler, D., Chopra, S., Frejd, F. Y., Yazaki, P. J. & Nestor, M. (2023). Enhanced Therapeutic Effects of 177Lu-DOTA-M5A in Combination with Heat Shock Protein 90 Inhibitor Onalespib in Colorectal Cancer Xenografts. Cancers, 15(17), Article ID 4239.
Open this publication in new window or tab >>Enhanced Therapeutic Effects of 177Lu-DOTA-M5A in Combination with Heat Shock Protein 90 Inhibitor Onalespib in Colorectal Cancer Xenografts
Show others...
2023 (English)In: Cancers, ISSN 2072-6694, Vol. 15, no 17, article id 4239Article in journal (Refereed) Published
Abstract [en]

Carcinoembryonic antigen (CEA) has emerged as an attractive target for theranostic applications in colorectal cancers (CRCs). In the present study, the humanized anti-CEA antibody hT84.66-M5A (M5A) was labeled with 177Lu for potential CRC therapy. Moreover, the novel combination of 177Lu-DOTA-M5A with the heat shock protein 90 inhibitor onalespib, suggested to mediate radiosensitizing properties, was assessed in vivo for the first time. M5A antibody uptake and therapeutic effects, alone or in combination with onalespib, were assessed in human CRC xenografts and visualized using SPECT/CT imaging. Although both 177Lu-DOTA-M5A and onalespib monotherapies effectively reduced tumor growth rates, the combination therapy demonstrated the most substantial impact, achieving a fourfold reduction in tumor growth compared to the control group. Median survival increased by 33% compared to 177Lu-DOTA-M5A alone, and tripled compared to control and onalespib groups. Importantly, combination therapy yielded comparable or superior effects to the double dose of 177Lu-DOTA-M5A monotherapy. 177Lu-DOTA-M5A increased apoptotic cell levels, indicating its potential to induce tumor cell death. These findings show promise for 177Lu-DOTA-M5A as a CRC therapeutic agent, and its combination with onalespib could significantly enhance treatment efficacy. Further in vivo studies are warranted to validate these findings fully and explore the treatment’s potential for clinical use.

Simple summary

Cancer treatment is hampered by the limitations of individual therapy modalities and the intricate nature of the disease. The administration of maximal monotherapy doses often leads to undesirable side effects and/or therapy resistance. As a result, there is a growing recognition of the importance of investigating combination therapy to effectively address these obstacles. In the present in vivo study, the therapeutic effects of combination therapy with the heat shock protein 90 inhibitor onalespib, a potential radiosensitizer, and 177Lu-DOTA-M5A for colorectal cancer (CRC) treatment were explored for the first time. The results demonstrated that the combination treatment was so effective that retained or even superior therapeutic effects could be achieved with only half the dose of administered 177Lu-DOTA-M5A, showing enhanced tumor growth suppression and increased apoptosis. Consequently, the combination therapy involving 177Lu-DOTA-M5A and onalespib constitutes a promising approach for treating metastatic CRCs. By enhancing therapeutic effects, minimizing therapy resistance, and reducing side effects, this approach has the potential to expand the patient population that can benefit from targeted treatment.

Place, publisher, year, edition, pages
MDPI, 2023
Keywords
radioimmunotherapy, combination therapy, carcinoembryonic antigen, 177Lu-DOTAM5A, HSP90 inhibitor onalespib
National Category
Radiology, Nuclear Medicine and Medical Imaging Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-510984 (URN)10.3390/cancers15174239 (DOI)001070060100001 ()37686514 (PubMedID)
Funder
Swedish Research Council, 2020-01377Swedish Cancer Society, CAN 21/1534Swedish Cancer Society, CAN 20 0191Swedish Childhood Cancer Foundation, PR2020-0023Swedish Childhood Cancer Foundation, TJ2021-0072Ulf Lundahls minnesfond
Available from: 2023-09-05 Created: 2023-09-05 Last updated: 2024-06-20Bibliographically approved
Berglund, H., Lundsten Salomonsson, S., Mohajershojai, T., Ferrer Gago, F. J., Lane, D. P. & Nestor, M. (2023). p53 stabilisation potentiates [177Lu]Lu-DOTATATE treatment in neuroblastoma xenografts. European Journal of Nuclear Medicine and Molecular Imaging
Open this publication in new window or tab >>p53 stabilisation potentiates [177Lu]Lu-DOTATATE treatment in neuroblastoma xenografts
Show others...
2023 (English)In: European Journal of Nuclear Medicine and Molecular Imaging, ISSN 1619-7070, E-ISSN 1619-7089Article in journal (Refereed) Epub ahead of print
Abstract [en]

Purpose

Molecular radiotherapy is a treatment modality that is highly suitable for targeting micrometastases and [177Lu]Lu-DOTATATE is currently being explored as a potential novel treatment option for high-risk neuroblastoma. p53 is a key player in the proapoptotic signalling in response to radiation-induced DNA damage and is therefore a potential target for radiosensitisation.

Methods

This study investigated the use of the p53 stabilising peptide VIP116 and [177Lu]Lu-DOTATATE, either alone or in combination, for treatment of neuroblastoma tumour xenografts in mice. Initially, the uptake of [177Lu]Lu-DOTATATE in the tumours was confirmed, and the efficacy of VIP116 as a monotherapy was evaluated. Subsequently, mice with neuroblastoma tumour xenografts were treated with placebo, VIP116, [177Lu]Lu-DOTATATE or a combination of both agents.

Results

The results demonstrated that monotherapy with either VIP116 or [177Lu]Lu-DOTATATE significantly prolonged median survival compared to the placebo group (90 and 96.5 days vs. 50.5 days, respectively). Notably, the combination treatment further improved median survival to over 120 days. Furthermore, the combination group exhibited the highest percentage of complete remission, corresponding to a twofold increase compared to the placebo group. Importantly, none of the treatments induced significant nephrotoxicity. Additionally, the therapies affected various molecular targets involved in critical processes such as apoptosis, hypoxia and angiogenesis.

Conclusion

In conclusion, the combination of VIP116 and [177Lu]Lu-DOTATATE presents a promising novel treatment approach for neuroblastoma. These findings hold potential to advance research efforts towards a potential cure for this vulnerable patient population.

Place, publisher, year, edition, pages
Springer, 2023
Keywords
Neuroblastoma, Molecular radiotherapy, p53, [177Lu]Lu-DOTATATE, Radiosensitisation
National Category
Radiology, Nuclear Medicine and Medical Imaging Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-517191 (URN)10.1007/s00259-023-06462-3 (DOI)001085247400001 ()37823909 (PubMedID)
Funder
Swedish Childhood Cancer Foundation, PR2020-0023Swedish Research Council, 2020-01377Swedish Cancer Society, CAN 20 0191Swedish Cancer Society, 2018/494Uppsala UniversitySwedish Childhood Cancer Foundation, TJ2021-0072Ulf Lundahls minnesfond
Note

De två första författarna delar förstaförfattarskapet

Available from: 2023-12-05 Created: 2023-12-05 Last updated: 2024-12-06Bibliographically approved
Fernandes, S. R. .., Mohajershojai, T., Lundsten, S., Sarmento, B., Tomé, J. P. .., Nestor, M. & Jha, P. (2023). Photoactive immunoconjugates for targeted photodynamic therapy of cancer. Journal of Photochemistry and Photobiology. B: Biology, 243, Article ID 112716.
Open this publication in new window or tab >>Photoactive immunoconjugates for targeted photodynamic therapy of cancer
Show others...
2023 (English)In: Journal of Photochemistry and Photobiology. B: Biology, ISSN 1011-1344, E-ISSN 1873-2682, Vol. 243, article id 112716Article in journal (Refereed) Published
Abstract [en]

Photodynamic therapy (PDT) has been used as an alternative or as a complement of conventional approaches for cancer treatment. In PDT, the reactive oxygen species (ROS) produced from the interaction between the photosensitizer (PS), visible light and molecular oxygen, kill malignant cells by triggering a cascade of cytotoxic reactions. In this process, the PS plays an extremely important role in the effectiveness of the therapy. In the present work, a new photoimmunoconjugate (PIC), based on cetuximab and the known third generation PS-glycophthalocyanine ZnPcGal4, was synthesized via reductive amination. The rationale behind this was the simultaneous cancer-associated specific targeting of PIC and photosensitization of targeted receptor positive cells. Varied reaction parameters and photodynamic conditions, such as PS concentrations and both type and intensities of light, were optimized. ZnPcGal4 showed significant photoactivity against EGFR expressing A431, EGFR-transfected HCT116 and HT29 cells when irradiated with white light of stronger intensity (38 mW/cm2). Similarly, the synthesized PICs-T1 and T2 also demonstrated photoactivity with high intensity white light. The best optimized PIC: sample 28 showed no precipitation and aggregation when inspected visually and analyzed through SE-HPLC. Fluorescence excitation of sample 28 and 125I-sample 28 radioconjugate (125I-PIC, 125I-radiolabeling yield ≥95%, determined with ITLC) at 660 nm showed presence of appended ZnPcGal4. In addition, simultaneous fluorescence and radioactivity detection of the 125I-PIC in serum and PBS (pH 7.4) for the longest incubated time point of 72 h, respectively, and superimposed signals thereof demonstrated ≥99% of loading and/or labeling yield, assuring overall stability of the PIC and corresponding PIC-radioconjugate w.r.t. both the appended ZnPcGal4 and bound-125I. Moreover, real-time binding analyses on EGFR-transfected HCT116 cells showed specific binding of 125I-PIC, suggesting no alternation in the binding kinetics of the mAb after appending it with ZnPcGal4. These results suggest dual potential applications of synthesized PICs both for PDT and radio-immunotherapy of cancer.

Place, publisher, year, edition, pages
Elsevier, 2023
National Category
Medicinal Chemistry
Identifiers
urn:nbn:se:uu:diva-508250 (URN)10.1016/j.jphotobiol.2023.112716 (DOI)001042154000001 ()37126865 (PubMedID)
Funder
Swedish Cancer Society, CAN 21/1534Swedish Cancer Society, CAN 20 0191Swedish Research Council, 2020-01377Swedish Childhood Cancer Foundation, PR2020-0023Swedish Childhood Cancer Foundation, TJ2021-0072Ulf Lundahls minnesfond
Available from: 2023-07-24 Created: 2023-07-24 Last updated: 2023-08-17Bibliographically approved
Mortensen, A., Berglund, H., Segerström, L., Walle, M., Hofström, C., Persson, H., . . . Nestor, M. (2023). Selection, characterization and in vivo evaluation of novel CD44v6-targeting antibodies for targeted molecular radiotherapy. Scientific Reports, 13, Article ID 20648.
Open this publication in new window or tab >>Selection, characterization and in vivo evaluation of novel CD44v6-targeting antibodies for targeted molecular radiotherapy
Show others...
2023 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 13, article id 20648Article in journal (Refereed) Published
Abstract [en]

Molecular radiotherapy combines the advantages of systemic administration of highly specific antibodies or peptides and the localized potency of ionizing radiation. A potential target for molecular radiotherapy is the cell surface antigen CD44v6, which is overexpressed in numerous cancers, with limited expression in normal tissues. The aim of the present study was to generate and characterize a panel of human anti-CD44v6 antibodies and identify a suitable candidate for future use in molecular radiotherapy of CD44v6-expressing cancers. Binders were first isolated from large synthetic phage display libraries containing human scFv and Fab antibody fragments. The antibodies were extensively analyzed through in vitro investigations of binding kinetics, affinity, off-target binding, and cell binding. Lead candidates were further subjected to in vivo biodistribution studies in mice bearing anaplastic thyroid cancer xenografts that express high levels of CD44v6. Additionally, antigen-dependent tumor uptake of the lead candidate was verified in additional xenograft models with varying levels of target expression. Interestingly, although only small differences were observed among the top antibody candidates in vitro, significant differences in tumor uptake and retention were uncovered in in vivo experiments. A high-affinity anti-CD44v6 lead drug candidate was identified, mAb UU-40, which exhibited favorable target binding properties and in vivo distribution. In conclusion, a panel of human anti-CD44v6 antibodies was successfully generated and characterized in this study. Through comprehensive evaluation, mAb UU-40 was identified as a promising lead candidate for future molecular radiotherapy of CD44v6-expressing cancers due to its high affinity, excellent target binding properties, and desirable in vivo distribution characteristics.

Place, publisher, year, edition, pages
Springer Nature, 2023
National Category
Cancer and Oncology Radiology, Nuclear Medicine and Medical Imaging
Identifiers
urn:nbn:se:uu:diva-521802 (URN)10.1038/s41598-023-47891-2 (DOI)001136085000078 ()38001360 (PubMedID)
Funder
Swedish Cancer Society
Available from: 2024-01-31 Created: 2024-01-31 Last updated: 2024-12-06Bibliographically approved
Mortensen, A., Imgenberg-Kreuz, J., Spiegelberg, D., Botling, J. & Nestor, M. (2023). Utilizing CD44v6 and V600EBRAF-mutation for in vitro targeted combination therapy of thyroid carcinomas. Heliyon, 9(12), Article ID e22594.
Open this publication in new window or tab >>Utilizing CD44v6 and V600EBRAF-mutation for in vitro targeted combination therapy of thyroid carcinomas
Show others...
2023 (English)In: Heliyon, E-ISSN 2405-8440, Vol. 9, no 12, article id e22594Article in journal (Refereed) Published
Abstract [en]

Aim: The aim of this study was to assess the feasibility of targeted therapy of thyroid carcinoma, first exploring potential targets BRAF, EGFR and CD44v6 in patient material through immunohistochemistry and mutation analysis.

Materials and methods: A patient cohort (n = 22) consisting of seven papillary (PTC), eight anaplastic (ATC) and seven follicular (FTC) thyroid carcinomas were evaluated. Additionally, eight thyroid carcinoma cells lines were analyzed for CD44v6-expression and sensitivity to the multi-kinase inhibitor sorafenib (Nexavar (R)), which targets numerous serine/threonine and tyrosine kinases, including the Raf family kinases. Targeted therapy using 131I-AbN44v6, a novel anti-CD44v6 antibody, and/or sorafenib was evaluated in 3D multicellular tumor spheroids.

Results: Of the two cell surface proteins, EGFR and CD44v6, the latter was overexpressed in >80 % of samples, while EGFR-expression levels were moderate at best in only a few samples. BRAF mutations were more common in PTC patient samples than in ATC samples, while FTC samples did not harbor BRAF mutations. CD44v6-expression levels in the thyroid carcinoma cell lines were more heterogenous compared to patient samples, while BRAF mutational status was in line with the original tumor type. Monotherapy in 3D multicellular ATC tumor spheroids with either 131I-AbN44v6 or sorafenib resulted in delayed spheroid growth. The combination of 131I-AbN44v6 and sorafenib was the most potent and resulted in significantly impaired spheroid growth.

Conclusion: This "proof of concept" targeted therapy study in the in vitro ATC 3D multicellular tumor spheroids indicated applicability of utilizing CD44v6 for molecular radiotherapy both as a monotherapy and in combination with sorafenib.

Place, publisher, year, edition, pages
Cell Press, 2023
Keywords
CD44v6, BRAF, Combination therapy, Thyroid cancer, Anaplastic thyroid cancer, targeted therapy, molecular radiotherapy
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-519104 (URN)10.1016/j.heliyon.2023.e22594 (DOI)001125311200001 ()38076095 (PubMedID)
Funder
Swedish Cancer SocietySwedish Research CouncilSwedish Society for Medical Research (SSMF)
Available from: 2024-01-04 Created: 2024-01-04 Last updated: 2024-01-04Bibliographically approved
Projects
Improving cancer treatment by combining radio-immunotargeting and p53 therapy [2013-03742_VR]; Uppsala UniversityNovel cancer-targeting strategies for improved molecular radiotherapy [2020-01377_VR]; Uppsala University; Publications
Ingelshed, K., Melssen, M. M., Kannan, P., Chandramohan, A., Partridge, A. W., Jiang, L., . . . Spiegelberg, D. (2024). MDM2/MDMX inhibition by Sulanemadlin synergizes with anti-Programmed Death 1 immunotherapy in wild-type p53 tumors. iScience, 27(6), Article ID 109862.
Advancing precision radiotherapy through novel cancer targeting strategies [2024-03447_VR]; Uppsala University
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0003-4662-3142

Search in DiVA

Show all publications