166 results on '"van der Veldt AAM"'
Search Results
2. P71 The Development of a Flexible and Easy to Tailor Disease Model to Estimate the Outcomes of Treatment Sequences in Advanced Melanoma by Combining Trial and Real-World Data
- Author
-
de Groot, S, primary, Blommestein, HM, additional, Leeneman, B, additional, Uyl-De, Groot CA, additional, Haanen, JBAG, additional, Suijkerbuijk, KPM, additional, Aarts, M.J.B., additional, van den Berkmortel, FWPJ, additional, Blank, CU, additional, Boers-Sonderen, MJ, additional, van den Eertwegh, AJM, additional, de Groot, JWB, additional, Hospers, GAP, additional, Kapiteijn, E, additional, de Meza, MM, additional, Piersma, D, additional, van Rijn, RS, additional, Stevense-den Boer, MAM, additional, van der Veldt, AAM, additional, Vreugdenhil, G, additional, Wouters, MWJM, additional, Franken, M, additional, and van Baal, PHM, additional
- Published
- 2022
- Full Text
- View/download PDF
3. POSC365 Health State Utilities of Advanced Melanoma Patients Treated in Clinical Practice in the Era of Novel Immuno- and Targeted Therapies
- Author
-
Franken, M, primary, de Groot, S, additional, van Dongen, A, additional, Leeneman, B, additional, Uyl-De Groot, CA, additional, Aarts, MJB, additional, van den Berkmortel, FWPJ, additional, Blank, CU, additional, Boers-Sonderen, MJ, additional, van den Eertwegh, AJM, additional, de Groot, JWB, additional, Haanen, JBAG, additional, Hospers, GAP, additional, Kapiteijn, E, additional, van Not, OJ, additional, Piersma, D, additional, van Rijn, RS, additional, Stevense-den Boer, MAM, additional, Suijkerbuijk, KPM, additional, van der Veldt, AAM, additional, Vreugdenhil, G, additional, Wouters, MWJM, additional, Versteegh, M, additional, and Blommestein, HM, additional
- Published
- 2022
- Full Text
- View/download PDF
4. POSB361 Quality of Life in Advanced Melanoma Patients in the Era of Novel Immuno- and Targeted Therapies
- Author
-
Franken, M, primary, Leeneman, B, additional, Aarts, M.J.B., additional, van den Berkmortel, FWPJ, additional, Blank, CU, additional, Boers-Sonderen, MJ, additional, van den Eertwegh, AJM, additional, de Groot, JWB, additional, Haanen, JBAG, additional, Hospers, GAP, additional, Kapiteijn, E, additional, van Not, OJ, additional, Piersma, D, additional, van Rijn, RS, additional, Stevense-den Boer, MAM, additional, Suijkerbuijk, KPM, additional, van der Veldt, AAM, additional, Vreugdenhil, G, additional, Wouters, MWJM, additional, van Dongen, A, additional, and Uyl-De Groot, CA, additional
- Published
- 2022
- Full Text
- View/download PDF
5. POSC366 Validity of the EQ-5D-3L and EQ-5D-5L in Advanced Melanoma
- Author
-
Franken, M, primary, van Dongen, A, additional, Leeneman, B, additional, Uyl-De Groot, CA, additional, Aarts, MJB, additional, van den Berkmortel, FWPJ, additional, Blank, CU, additional, Boers-Sonderen, MJ, additional, van den Eertwegh, AJM, additional, de Groot, JWB, additional, Haanen, JBAG, additional, Hospers, GAP, additional, Kapiteijn, E, additional, de Meza, MM, additional, Piersma, D, additional, van Rijn, RS, additional, Stevense-den Boer, MAM, additional, Suijkerbuijk, KPM, additional, van der Veldt, AAM, additional, Vreugdenhil, G, additional, Wouters, MWJM, additional, Blommestein, HM, additional, and Versteegh, M, additional
- Published
- 2022
- Full Text
- View/download PDF
6. The role of local therapy in the treatment of solitary melanoma progression on immune checkpoint inhibition: A multicentre retrospective analysis
- Author
-
Versluis, JM, Hendriks, AM, Weppler, AM, Brown, LJ, de Joode, K, Suijkerbuijk, KPM, Zimmer, L, Kapiteijn, EW, Allayous, C, Johnson, DB, Hepner, A, Mangana, J, Bhave, P, Jansen, YJL, Trojaniello, C, Atkinson, V, Storey, L, Lorigan, P, Ascierto, PA, Neyns, B, Haydon, A, Menzies, AM, Long, G, Lebbe, C, van der Veldt, AAM, Carlino, MS, Sandhu, S, van Tinteren, H, de Vries, EGE, Blank, CU, Jalving, M, Versluis, JM, Hendriks, AM, Weppler, AM, Brown, LJ, de Joode, K, Suijkerbuijk, KPM, Zimmer, L, Kapiteijn, EW, Allayous, C, Johnson, DB, Hepner, A, Mangana, J, Bhave, P, Jansen, YJL, Trojaniello, C, Atkinson, V, Storey, L, Lorigan, P, Ascierto, PA, Neyns, B, Haydon, A, Menzies, AM, Long, G, Lebbe, C, van der Veldt, AAM, Carlino, MS, Sandhu, S, van Tinteren, H, de Vries, EGE, Blank, CU, and Jalving, M
- Abstract
INTRODUCTION: In patients with metastatic melanoma, progression of a single tumour lesion (solitary progression) after response to immune checkpoint inhibition (ICI) is increasingly treated with local therapy. We evaluated the role of local therapy for solitary progression in melanoma. PATIENTS AND METHODS: Patients with metastatic melanoma treated with ICI between 2010 and 2019 with solitary progression as first progressive event were included from 17 centres in 9 countries. Follow-up and survival are reported from ICI initiation. RESULTS: We identified 294 patients with solitary progression after stable disease in 15%, partial response in 55% and complete response in 30%. The median follow-up was 43 months; the median time to solitary progression was 13 months, and the median time to subsequent progression after treatment of solitary progression (TTSP) was 33 months. The estimated 3-year overall survival (OS) was 79%; median OS was not reached. Treatment consisted of systemic therapy (18%), local therapy (36%), both combined (42%) or active surveillance (4%). In 44% of patients treated for solitary progression, no subsequent progression occurred. For solitary progression during ICI (n = 143), the median TTSP was 29 months. Both TTSP and OS were similar for local therapy, ICI continuation and both combined. For solitary progression post ICI (n = 151), the median TTSP was 35 months. TTSP was higher for ICI recommencement plus local therapy than local therapy or ICI recommencement alone (p = 0.006), without OS differences. CONCLUSION: Almost half of patients with melanoma treated for solitary progression after initial response to ICI had no subsequent progression. This study suggests that local therapy can benefit patients and is associated with favourable long-term outcomes.
- Published
- 2021
7. Genomic and Transcriptomic Characterization of Metastatic Urothelial Carcinoma
- Author
-
Nakauma, A, primary, Rijnders, M, additional, van Riet, J, additional, van der Heijden, MS, additional, Voortman, J, additional, Cuppen, E, additional, Mehra, N, additional, van Wilpe, S, additional, Oosting, S, additional, Zwarthoff, EC, additional, de Wit, R, additional, van der Veldt, AAM, additional, van de Werken, HJG, additional, Lolkema, MPJ, additional, and Boormans, JL, additional
- Published
- 2020
- Full Text
- View/download PDF
8. Effect of Bisphosphonates on Skeletal Related Events in Long Bone Metastases of Renal Cell Carcinoma: A Systematic Review.
- Author
-
van Broekhoven, DLM, Dootjes, EW, van der Veldt, AAM, Zillikens, MC, and van Oldenrijk, J
- Abstract
Bone metastases (BMs) in patients with renal cell carcinoma (RCC) are lytic lesions which are prone to skeletal related events (SREs) such as (pending) pathological fractures or bone pain requiring radiotherapy or surgery. The aim of this review is to assess whether the use of bisphosphonates in patients with RCC and BMs in the long bones results in reduced SRE rate.
- Published
- 2023
- Full Text
- View/download PDF
9. An Overview of Liver Directed Locoregional Therapies.
- Author
-
Höppener DJ, Grünhagen DJ, Eggermont AMM, van der Veldt AAM, and Verhoef C
- Subjects
- Humans, Uveal Neoplasms therapy, Uveal Neoplasms pathology, Skin Neoplasms therapy, Skin Neoplasms pathology, Liver pathology, Combined Modality Therapy, Liver Neoplasms secondary, Liver Neoplasms therapy, Melanoma therapy, Melanoma pathology
- Abstract
An overview of all liver-directed locoregional therapies, including surgical resection for melanoma liver metastases (MLMs), is provided. MLM patients are divided by their primary melanoma location; cutaneous, uvea (eye), and mucosal melanoma. If patients with isolated cutaneous MLMs are considered for surgical resection, treatment with systemic therapy should be part of the treatment course. For uveal MLMs, complete surgical or ablative treatment of all MLMs suggests superior results compared with other liver-directed or systemic therapies, based on current evidence, no recommendations for any liver-directed regional therapy in the treatment of mucosal MLMs can be made., Competing Interests: Disclosure Authors have nothing to disclose., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
10. First real-world clinical experience with [ 177 Lu]Lu-PSMA-I&T in patients with metastatic castration-resistant prostate cancer beyond VISION and TheraP criteria.
- Author
-
Ling SW, de Lussanet de la Sablonière Q, Ananta M, de Blois E, Koolen SLW, Drexhage RC, Hofland J, Robbrecht DGJ, van der Veldt AAM, Verburg FA, and Brabander T
- Abstract
Purpose: To report real-world clinical experience with [
177 Lu]Lu-PSMA-I&T targeted radionuclide therapy (TRT) in patients with metastatic castration-resistant prostate cancer (mCRPC) in a single tertiary referral university hospital., Methods: Patients with mCRPC who were treated with [177 Lu]Lu-PSMA-I&T TRT as standard of care between February 2022 and August 2023 were included in this retrospective study. Patients were treated with a maximum of six cycles with a fixed activity of 7.4 GBq/100µg [177 Lu]Lu-PSMA-I&T per cycle., Results: 50 patients with mCRPC were included, of them 84% had prior therapy with two lines of taxane-based chemotherapy treated and at least one line of androgen receptor signaling inhibitor. A total of 126 cycles with a median of 2 cycles (IQR 1-6) [177 Lu]Lu-PSMA-I&T were administered per patient. PSA declines of ≥ 50% and ≥ 70% were achieved in 16% and 10% of the patients, respectively. Radiological response was achieved in 11% of the patients. In total, 68 treatment-related Adverse Events (TRAEs) were observed, mainly grade 1-2 in 88% of cases. Grade 3/4 TRAEs were observed in 12% of cases. No grade 3 or higher xerostomia was reported. Median progression-free survival was 7.7 months (95% CI 4.0-11.3) and median overall survival was 8.1 months (95% CI 5.0-11.3)., Conclusion: In heavily pretreated patients with mCRPC, treatment of [177 Lu]Lu-PSMA-I&T TRT is well tolerated and safe, but real-world efficacy of [177 Lu]Lu-PSMA appears lower compared to data from recent phase-3 clinical trials using a different radioligand [177 Lu]Lu-PSMA-617. Further studies may show whether patients with mCRPC benefit more from [177 Lu]Lu-PSMA when initiated at an earlier stage of treatment., Competing Interests: Declarations. Ethics approval: This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of the Erasmus Medical Center (15 March 2024/MEC-2024-0104). Consent to participate: Informed consent from all individual participants included in the study was waived by the Ethics Committee of the Erasmus Medical Center under special circumstances. Competing interests: The authors have no relevant financial or non-financial interests to disclose., (© 2025. The Author(s).)- Published
- 2025
- Full Text
- View/download PDF
11. Bioequivalence of alternative pembrolizumab dosing regimens: current practice and future perspectives.
- Author
-
Malmberg R, Agema BC, Hofman MM, Oosterveld S, Bins S, Dumoulin DW, Joosse A, Aerts JGJV, Debets R, Koch BCP, van der Veldt AAM, van Leeuwen RWF, and Mathijssen RHJ
- Published
- 2025
- Full Text
- View/download PDF
12. Impact of personalized response-directed surgery and adjuvant therapy on survival after neoadjuvant immunotherapy in stage III melanoma: Comparison of 3-year data from PRADO and OpACIN-neo.
- Author
-
Reijers ILM, Menzies AM, Lopez-Yurda M, Versluis JM, Rozeman EA, Saw RPM, van Houdt WJ, Kapiteijn E, van der Veldt AAM, Suijkerbuijk KPM, Eriksson H, Hospers GAP, Klop WMC, Torres Acosta A, Grijpink-Ongering L, Gonzalez M, van der Wal A, Al-Mamgani A, Spillane AJ, Scolyer RA, van de Wiel BA, van Akkooi ACJ, Long GV, and Blank CU
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Chemotherapy, Adjuvant, Skin Neoplasms mortality, Skin Neoplasms therapy, Skin Neoplasms pathology, Skin Neoplasms drug therapy, Skin Neoplasms immunology, Lymph Node Excision, Immune Checkpoint Inhibitors therapeutic use, Adult, Immunotherapy methods, Nivolumab therapeutic use, Precision Medicine methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Melanoma mortality, Melanoma therapy, Melanoma pathology, Melanoma drug therapy, Melanoma immunology, Neoadjuvant Therapy mortality, Neoadjuvant Therapy methods, Neoplasm Staging, Ipilimumab therapeutic use
- Abstract
Background: Pathologic response following neoadjuvant immune checkpoint blockade (ICB) in stage III melanoma serves as a surrogate marker for long-term outcomes. This may support more personalized, response-directed treatment strategies., Methods: The OpACIN-neo and PRADO trials were phase 2 studies evaluating neoadjuvant treatment with ipilimumab and nivolumab in stage III melanoma. In OpACIN-neo, all patients underwent therapeutic lymph node dissection (TLND) without subsequent adjuvant therapy. In contrast, PRADO explored a response- directed strategy, where patients achieving a major pathologic response (MPR) omitted TLND and adjuvant therapy, while those without a pathologic response (pNR) received TLND and adjuvant therapy. Here, we provide a descriptive post-hoc comparison of 3-year survival outcomes between the non-personalized approach in OpACIN-neo and the response-directed approach in PRADO., Results: For patients who achieved an MPR, the 3-year recurrence-free survival (RFS) was 93 % for those without TLND versus 96 % for those with TLND (log-rank p = 0.47), and distant metastasis-free survival (DMFS) was 98 % compared to 96 % (log-rank p = 0.49), respectively. For patients with pNR, 3-year RFS rates were 64 % for those receiving adjuvant systemic therapy and 35 % for patients without (log-rank p = 0.10). DMFS rates were 70 % versus 52 % (log-rank p = 0.24), respectively., Conclusions: These data suggest that TLND and adjuvant therapy may be safely omitted in most patients achieving an MPR, while adjuvant systemic therapy following TLND appears to improve RFS and DMFS in patients with pNR. Although these results are hypothesis-generating and require further validation, they offer a potential foundation for developing personalized neoadjuvant immunotherapy approaches., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: No author has received financial support for the work on this manuscript. I.L.M.R reports financial interest in Signature Oncology. A.M.M. has served on advisory boards for Bristol Myers Squibb (BMS), Merck Sharp & Dohme (MSD), Novartis, Roche, Pierre Fabre and QBiotics. R.P.M.S. has received honoraria for advisory board participation from MSD, Novartis and Qbiotics and speaking honoraria from BMS and Novartis. W.J.v.H. reports an advisory role for Amgen, BMS, and Sanofi. E.K.A. received honoraria for consultancy/advisory relationships (all paid to the institute) from BMS, Novartis, Merck, Lilly, Immunocore and Pierre Fabre, and received research grants not related to this paper from BMS, Delcath, Pierre-Fabre and Novartis. A.A.M.v.d.V. received compensation for advisory roles and honoraria (all paid to the institute) from BMS, MSD, Merck, Roche, Eisai, Pfizer, Sanofi, Novartis, Pierre Fabre, and Ipsen. K.P.M.S. received compensation for advisory roles and honoraria (all paid to the institute) from BMS, MSD, Novartis, Pierre Fabre, Sairopa and Abbvie and received research funding from Philips, Novartis, TigaTx and BMS. H.E. has served on advisory boards for BMS, Novartis and Pierre Fabre Nordic, and received project funding from SkyLineDx. G.A.P.H. received compensation for consulting and advisory roles (all paid to the institute) from Amgen, Roche, MSD, BMS, Pfizer, Novartis and Pierre Fabre, and received research grants (paid to the institute) from BMS and Seerave. R.A.S. has received fees for professional services from MetaOptima Technology Inc., F. Hoffmann-La Roche, Evaxion, Provectus Biopharmaceuticals Australia, Qbiotics, Novartis, MSD, NeraCare, Amgen, BMS, Myriad Genetics and GlaxoSmithKline. B.A.v.d.W. has served on the advisory board for BMS. A.C.J.v.A. has served on advisory boards and received consultancy honoraria for Amgen, BMS, Neracare, Novartis, MSD Merck, Merck-Pfizer, Pierre Fabre, Provectus, Sanofi, Sirius Medical and 4SC, and received research grants (all paid to the institute) from Amgen and Merck-Pfizer. G.V.L. is consultant advisor for Agenus, Amgen, Array Biopharma, AstraZeneca, Boehringer Ingelheim, BMS, Evaxion, Hexal AG (Sandoz Company), Highlight Therapeutics, Innovent Biologics, MSD, Novartis, Oncosec, PHMR Ltd, Pierre Fabre, Provectus, QBiotics and Regeneron Pharmaceuticals. C.U.B. reports receiving compensation for advisory roles from BMS, MSD, Roche, Novartis, GlaxoSmithKline, AstraZeneca, Pfizer, Eli Lilly, GenMab, Pierre Fabre and Third Rock Ventures and receiving research funding from BMS, MSD, Novartis, 4SC and NanoString. Furthermore, C.U.B. reports to be co-founder of Immagene BV and Signature Oncology. All compensations and funding for C.U.B. were paid to the institute, except for Third Rock Ventures, Immagene and Signature Oncology. The other authors declare no conflicts of interest., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
13. Corrigendum to "Adjuvant immunotherapy in older patients with stage III and resected stage IV melanoma: Toxicity and recurrence-free survival outcomes from the Dutch melanoma treatment registry" [Eur. J. Cancer 212, 2024, 115056].
- Author
-
Özkan A, Kapiteijn E, van den Bos F, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Bloem M, Blokx WAM, Boers-Sonderen MJ, Bonenkamp JJ, van den Eertwegh AJM, de Groot JWB, Haanen JB, Holtslag CE, Hospers GAP, Piersma D, van Rijn RS, Stevense-den Boer AM, Suijkerbuijk KPM, van der Veldt AAM, Vreugdenhil G, Wouters MWJM, Portielje JEA, and de Glas NA
- Published
- 2024
- Full Text
- View/download PDF
14. Local administration of immunotherapy for patients with skin cancer: A systematic review.
- Author
-
Janssen JC, van Dijk B, Hoeijmakers LL, Grünhagen DJ, Bramer WM, Verhoef C, de Gruijl TD, Blank CU, and van der Veldt AAM
- Subjects
- Humans, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell immunology, Carcinoma, Squamous Cell therapy, Injections, Intralesional, Skin Neoplasms drug therapy, Skin Neoplasms immunology, Skin Neoplasms therapy, Immunotherapy methods, Immune Checkpoint Inhibitors adverse effects, Immune Checkpoint Inhibitors administration & dosage, Immune Checkpoint Inhibitors therapeutic use, Melanoma drug therapy, Melanoma immunology, Melanoma therapy
- Abstract
Since the introduction of immune checkpoint inhibitors (ICIs) targeting PD-1 and CTLA-4 receptors, survival has improved significantly for patients with irresectable and metastatic skin cancer, including cutaneous squamous cell cancer and melanoma. However, systemic administration of these drugs is associated with immune related adverse events (irAEs), which can be severe, irreversible and even fatal. To reduce the risk of irAEs associated with systemic exposure to immunotherapeutic drugs, local administration of low doses could be considered. This systematic review provides an overview of early phase clinical trials with drugs that are currently under investigation for intratumoral administration in patients with melanoma and non-melanoma skin cancer., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: [T.D. de Gruijl: Consultancy/advisory board fees (all paid to institute): LAVA Therapeutics, Mendus, GE Health; research support: Idera Pharmaceuticals/Aceragen, BMS, Pfizer, Astra Zeneca; holds stocks from LAVA Therapeutics. C. Blank: Consultancy/advisory board fees (all paid to the institute): Bristol Muyers Squibb, F. Hoffman-La Roche, Merck; Research grants (all paid to the institute) 4SC, Nanostring Technologies; Advisory board fee (individual): Third Rock Ventures A.A.M. van der Veldt: consultancy fees (all paid to the institute): BMS, Eisai, Ipsen, MSD, Novartis, Pfizer, Pierre Fabre, Roche, Sanofi. J.C. Janssen none, B. van Dijk none, D.J. Grünhagen none, L.L. Hoeijmakers none, W.M. Bramer none, C. Verhoef none]., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Neoadjuvant Nivolumab and Ipilimumab in Resectable Stage III Melanoma.
- Author
-
Blank CU, Lucas MW, Scolyer RA, van de Wiel BA, Menzies AM, Lopez-Yurda M, Hoeijmakers LL, Saw RPM, Lijnsvelt JM, Maher NG, Pulleman SM, Gonzalez M, Torres Acosta A, van Houdt WJ, Lo SN, Kuijpers AMJ, Spillane A, Klop WMC, Pennington TE, Zuur CL, Shannon KF, Seinstra BA, Rawson RV, Haanen JBAG, Ch'ng S, Naipal KAT, Stretch J, van Thienen JV, Rtshiladze MA, Wilgenhof S, Kapoor R, Meerveld-Eggink A, Grijpink-Ongering LG, van Akkooi ACJ, Reijers ILM, Gyorki DE, Grünhagen DJ, Speetjens FM, Vliek SB, Placzke J, Spain L, Stassen RC, Amini-Adle M, Lebbé C, Faries MB, Robert C, Ascierto PA, van Rijn R, van den Berkmortel FWPJ, Piersma D, van der Westhuizen A, Vreugdenhil G, Aarts MJB, Stevense-den Boer MAM, Atkinson V, Khattak M, Andrews MC, van den Eertwegh AJM, Boers-Sonderen MJ, Hospers GAP, Carlino MS, de Groot JB, Kapiteijn E, Suijkerbuijk KPM, Rutkowski P, Sandhu S, van der Veldt AAM, and Long GV
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Chemotherapy, Adjuvant methods, Chemotherapy, Adjuvant statistics & numerical data, Disease-Free Survival, Kaplan-Meier Estimate, Progression-Free Survival, Young Adult, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Ipilimumab administration & dosage, Ipilimumab adverse effects, Ipilimumab therapeutic use, Melanoma mortality, Melanoma pathology, Melanoma therapy, Neoadjuvant Therapy methods, Neoadjuvant Therapy statistics & numerical data, Neoplasm Staging, Nivolumab therapeutic use, Nivolumab adverse effects, Nivolumab administration & dosage, Skin Neoplasms mortality, Skin Neoplasms pathology, Skin Neoplasms therapy
- Abstract
Background: In phase 1-2 trials in patients with resectable, macroscopic stage III melanoma, neoadjuvant immunotherapy was more efficacious than adjuvant immunotherapy., Methods: In this phase 3 trial, we randomly assigned patients with resectable, macroscopic stage III melanoma to two cycles of neoadjuvant ipilimumab plus nivolumab followed by surgery or surgery followed by 12 cycles of adjuvant nivolumab. Only patients in the neoadjuvant group with a partial response or nonresponse received adjuvant treatment. The primary end point was event-free survival., Results: A total of 423 patients underwent randomization. At a median follow-up of 9.9 months, the estimated 12-month event-free survival was 83.7% (99.9% confidence interval [CI], 73.8 to 94.8) in the neoadjuvant group and 57.2% (99.9% CI, 45.1 to 72.7) in the adjuvant group. The difference in restricted mean survival time was 8.00 months (99.9% CI, 4.94 to 11.05; P<0.001; hazard ratio for progression, recurrence, or death, 0.32; 99.9% CI, 0.15 to 0.66). In the neoadjuvant group, 59.0% of patients had a major pathological response, 8.0% had a partial response, 26.4% had a nonresponse (>50% residual viable tumor), and 2.4% had progression; in 4.2%, surgery had not yet been performed or was omitted. The estimated 12-month recurrence-free survival was 95.1% in patients in the neoadjuvant group who had a major pathological response, 76.1% among those with a partial response, and 57.0% among those with a nonresponse. Adverse events of grade 3 or higher that were related to systemic treatment occurred in 29.7% of patients in the neoadjuvant group and in 14.7% in the adjuvant group., Conclusions: Among patients with resectable, macroscopic stage III melanoma, neoadjuvant ipilimumab plus nivolumab followed by surgery and response-driven adjuvant therapy resulted in longer event-free survival than surgery followed by adjuvant nivolumab. (Funded by Bristol Myers Squibb and others; NADINA ClinicalTrials.gov number, NCT04949113.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
16. Comprehensive characterization of circulating tumor cells and cell-free DNA in patients with metastatic melanoma.
- Author
-
Bos MK, Kraan J, Starmans MPA, Helmijr JCA, Verschoor N, De Jonge MJA, Joosse A, van der Veldt AAM, Te Boekhorst PAW, Martens JWM, Sleijfer S, and Wilting SM
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Neoplasm Metastasis, Biomarkers, Tumor blood, Biomarkers, Tumor genetics, Adult, Mutation, DNA Methylation genetics, Cell-Free Nucleic Acids blood, Cell-Free Nucleic Acids genetics, Aged, 80 and over, Aneuploidy, Liquid Biopsy, Neoplastic Cells, Circulating pathology, Neoplastic Cells, Circulating metabolism, Melanoma genetics, Melanoma blood, Melanoma pathology, Circulating Tumor DNA blood, Circulating Tumor DNA genetics
- Abstract
Advances in therapeutic approaches for melanoma urge the need for biomarkers that can identify patients at risk for recurrence and to guide treatment. The potential use of liquid biopsies in identifying biomarkers is increasingly being recognized. Here, we present a head-to-head comparison of several techniques to analyze circulating tumor cells (CTCs) and cell-free DNA (cfDNA) in 20 patients with metastatic melanoma. In this study, we investigated whether diagnostic leukapheresis (DLA) combined with multimarker flow cytometry (FCM) increased the detection of CTCs in blood compared to the CellSearch platform. Additionally, we characterized cfDNA at the level of somatic mutations, extent of aneuploidy and genome-wide DNA methylation. Both CTCs and cfDNA measures were compared to tumor markers and extracranial tumor burden on radiological imaging. Compared to the CellSearch method applied on peripheral blood, DLA combined with FCM increased the proportion of patients with detectable CTCs from 35% to 70% (P = 0.06). However, the median percentage of cells that could be recovered by the DLA procedure was 29%. Alternatively, cfDNA mutation and methylation analysis detected tumor load in the majority of patients (90% and 93% of samples successfully analyzed, respectively). The aneuploidy score was positive in 35% of all patients. From all tumor measurements in blood, lactate dehydrogenase (LDH) levels were significantly correlated to variant allele frequency (P = 0.004). Furthermore, the presence of CTCs in DLA was associated with tumor burden (P < 0.001), whereas the presence of CTCs in peripheral blood was associated with number of lesions on radiological imaging (P < 0.001). In conclusion, DLA tended to increase the proportion of patients with detectable CTCs but was also associated with low recovery. Both cfDNA and CTCs were correlated with clinical parameters such as LDH levels and extracranial tumor burden., (© 2024 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
17. Adjuvant immunotherapy in older patients with stage III and resected stage IV melanoma: Toxicity and recurrence-free survival outcomes from the Dutch melanoma treatment registry.
- Author
-
Özkan A, Kapiteijn E, van den Bos F, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Bloem M, Blokx WAM, Boers-Sonderen MJ, Bonenkamp JJ, van den Eertwegh AJM, de Groot JWB, Haanen JB, Holtslag CE, Hospers GAP, Piersma D, van Rijn RS, Stevense-den Boer AM, Suijkerbuijk KPM, van der Veldt AAM, Vreugdenhil G, Wouters MWJM, Portielje JEA, and de Glas NA
- Subjects
- Humans, Male, Female, Aged, Netherlands epidemiology, Aged, 80 and over, Chemotherapy, Adjuvant adverse effects, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors adverse effects, Immunotherapy adverse effects, Immunotherapy methods, Neoplasm Recurrence, Local, Age Factors, Melanoma mortality, Melanoma pathology, Melanoma drug therapy, Registries, Skin Neoplasms mortality, Skin Neoplasms pathology, Skin Neoplasms drug therapy, Neoplasm Staging
- Abstract
Background: Adjuvant anti-PD-1 therapy improves relapse free survival in stage III melanoma, but also leads to immune-related adverse events (irAEs). Older patients are of particular interest due to comorbidities and frailty, which may impact their ability to tolerate irAEs and benefit from anti-PD-1 therapy. This study aimed to explore associations between clinical parameters and the occurrence of grade ≥ 3 irAEs and recurrence-free survival (RFS) in older patients with radically resected stage III/IV cutaneous melanoma treated with adjuvant anti-PD-1 therapy., Methods: Patients aged ≥ 65 with resected stage III/IV cutaneous melanoma treated with adjuvant anti-PD-1 therapy between 2018 and 2022 were selected using real-world data from the nationwide Dutch Melanoma Treatment Registry (DMTR). A univariate and multivariable logistic regression was used to compare determinants of grade ≥ 3 irAEs, and univariate and multivariable Cox-proportional hazard models were fitted to identify factors influencing RFS., Results: The study included 885 patients, with 280 aged 75 and older. The incidence of grade ≥ 3 irAEs was 15.5 % in the 65-74 age group and 13.9 % in the ≥ 75 age group. No significant correlation was found between age and grade ≥ 3 irAEs. However, an increasing number of comorbidities was associated with a higher risk of grade ≥ 3 irAEs (multivariable analyses: OR 1.83, 95 % C.I. 0.99-3.40). The 1-year RFS rate of 80.0 % of this study was comparable to those reported in previous registration trials and real-world data. Having ≥ 3 comorbidities was significantly associated with a decrease in RFS (HR: 1.68, 95 % C.I. 1.15-2.44)., Conclusion: Older patients had similar benefit of adjuvant immunotherapy compared to older subgroups in previous trials. However, patients with multiple comorbidities were at increased risk of grade ≥ 3 irAEs and had a lower RFS. This should be considered when deciding upon adjuvant treatment., Competing Interests: Declaration of Competing Interest All remaining authors have declared no conflicts of interest., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Adjuvant treatment with anti-PD-1 in acral melanoma: A nationwide study.
- Author
-
Bloem M, van Not OJ, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Blokx WAM, Boers-Sonderen MJ, Bonenkamp JJ, de Groot JB, Haanen JB, Hospers GAP, Kapiteijn EW, de Meza MM, Piersma D, van Rijn RS, Stevense-den Boer MAM, van der Veldt AAM, Vreugdenhil G, van den Eertwegh AJM, Suijkerbuijk KPM, and Wouters MWJM
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Chemotherapy, Adjuvant methods, Prospective Studies, Adult, Mutation, Netherlands epidemiology, Aged, 80 and over, Melanoma, Cutaneous Malignant, Registries, Programmed Cell Death 1 Receptor antagonists & inhibitors, Melanoma drug therapy, Melanoma genetics, Melanoma mortality, Melanoma pathology, Immune Checkpoint Inhibitors therapeutic use, Skin Neoplasms drug therapy, Skin Neoplasms pathology, Skin Neoplasms genetics, Skin Neoplasms mortality
- Abstract
Previous studies demonstrated limited efficacy of immune checkpoint inhibitors in unresectable acral melanoma (AM); it remains unclear how this translates to the adjuvant setting. This study investigates clinical outcomes of acral compared to cutaneous melanoma (CM) patients treated with adjuvant anti-PD-1 after complete resection. All stages III-IV AM and CM patients receiving adjuvant anti-PD-1 after complete resection between 2018 and 2022 were included from the prospective nationwide Dutch Melanoma Treatment Registry. We analyzed recurrence-free survival (RFS), distant metastasis-free survival (DMFS), and overall survival (OS). A multivariable Cox regression analysis of RFS was performed to adjust for potential confounders. We included 1958 (86 AM and 1872 CM) patients. At baseline, AM patients more frequently had KIT mutations, higher disease stages, and Eastern Cooperative Oncology Group Performance Status, and fewer BRAF and NRAS mutations. Median RFS was 14.8 months (95% confidence interval [CI]: 11.5-29.3) in AM and 37.4 months (95% CI: 34.6 to not reached) in CM (p = .002). After correcting for potential confounders, AM remained associated with a higher risk of recurrence (HR
adj 1.53; 95% CI: 1.07-2.17; p = .019). Two-year DMFS tended to be worse for AM than for CM: 64.5% versus 79.7% (p = .050). Two-year OS was significantly lower in AM (71.5% vs. 84.3%; p = .027). The results of this study suggest a poorer outcome of adjuvant-treated AM compared to CM. Studies assessing the added value of adjuvant treatment in AM are needed. Future research should investigate alternative treatment strategies to improve outcomes of high-risk AM., (© 2024 The Author(s). International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)- Published
- 2024
- Full Text
- View/download PDF
19. The development of brain metastases in patients with different therapeutic strategies for metastatic renal cell cancer.
- Author
-
Derks SHAE, van der Meer EL, Joosse A, de Jonge MJA, Slagter C, Schouten JW, Hoop EO, Smits M, van den Bent MJ, Jongen JLM, and van der Veldt AAM
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Quality of Life, Retrospective Studies, Netherlands epidemiology, Carcinoma, Renal Cell secondary, Carcinoma, Renal Cell pathology, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell therapy, Brain Neoplasms secondary, Brain Neoplasms therapy, Kidney Neoplasms pathology
- Abstract
A diagnosis of brain metastasis (BM) significantly affects quality of life in patients with metastatic renal cell cancer (mRCC). Although systemic treatments have shown efficacy in mRCC, active surveillance (AS) is still commonly used in clinical practice. In this single-center cohort study, we assessed the impact of different initial treatment strategies for metastatic RCC (mRCC) on the development of BM. All consecutive patients diagnosed with mRCC between 2011 and 2022 were included at the Erasmus MC Cancer Institute, the Netherlands, and a subgroup of patients with BM was selected. In total, 381 patients with mRCC (ECM, BM, or both) were identified. Forty-six patients had BM of whom 39 had metachronous BM (diagnosed ≥1 month after ECM). Twenty-five (64.1%) of these 39 patients with metachronous BM had received prior systemic treatment for ECM and 14 (35.9%) patients were treatment naive at BM diagnosis. The median BM-free survival since ECM diagnosis was significantly longer (p = .02) in previously treated patients (29.0 [IQR 12.6-57.0] months) compared to treatment naive patients (6.8 [IQR 1.0-7.0] months). In conclusion, patients with mRCC who received systemic treatment for ECM prior to BM diagnosis had a longer BM-free survival as compared to treatment naïve patients. These results emphasize the need for careful evaluation of treatment strategies, and especially AS, for patients with mRCC., (© 2024 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2024
- Full Text
- View/download PDF
20. mRNA-1273 vaccination induces polyfunctional memory CD4 and CD8 T cell responses in patients with solid cancers undergoing immunotherapy or/and chemotherapy.
- Author
-
Gangaev A, van Sleen Y, Brandhorst N, Hoefakker K, Prajapati B, Singh A, Boerma A, van der Heiden M, Oosting SF, van der Veldt AAM, Hiltermann TJN, GeurtsvanKessel CH, Dingemans AC, Smit EF, de Vries EGE, Haanen JBAG, Kvistborg P, and van Baarle D
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Immunotherapy methods, Adult, COVID-19 Vaccines immunology, Vaccination, Spike Glycoprotein, Coronavirus immunology, Immunologic Memory, Neoplasms immunology, Neoplasms therapy, Neoplasms drug therapy, CD8-Positive T-Lymphocytes immunology, COVID-19 immunology, COVID-19 prevention & control, CD4-Positive T-Lymphocytes immunology, SARS-CoV-2 immunology, 2019-nCoV Vaccine mRNA-1273 immunology, Memory T Cells immunology
- Abstract
Introduction: Research has confirmed the safety and comparable seroconversion rates following SARS-CoV-2 vaccination in patients with solid cancers. However, the impact of cancer treatment on vaccine-induced T cell responses remains poorly understood., Methods: In this study, we expand on previous findings within the VOICE trial by evaluating the functional and phenotypic composition of mRNA-1273-induced T cell responses in patients with solid tumors undergoing immunotherapy, chemotherapy, or both, compared to individuals without cancer. We conducted an ELISpot analysis on 386 participants to assess spike-specific T cell responses 28 days after full vaccination. Further in-depth characterization of using flow cytometry was performed on a subset of 63 participants to analyze the functional phenotype and differentiation state of spike-specific T cell responses., Results: ELISpot analysis showed robust induction of spike-specific T cell responses across all treatment groups, with response rates ranging from 75% to 80%. Flow cytometry analysis revealed a distinctive cytokine production pattern across cohorts, with CD4 T cells producing IFNγ, TNF, and IL-2, and CD8 T cells producing IFNγ, TNF, and CCL4. Variations were observed in the proportion of monofunctional CD4 T cells producing TNF, particularly higher in individuals without cancer and patients treated with chemotherapy alone, while those treated with immunotherapy or chemoimmunotherapy predominantly produced IFNγ. Despite these differences, polyfunctional spike-specific memory CD4 and CD8 T cell responses were comparable across cohorts. Notably, immunotherapy-treated patients exhibited an expansion of spike-specific CD4 T cells with a terminally differentiated effector memory phenotype., Discussion: These findings demonstrate that systemic treatment in patients with solid tumors does not compromise the quality of polyfunctional mRNA-1273-induced T cell responses. This underscores the importance of COVID-19 vaccination in patients with solid cancers undergoing systemic treatment., Competing Interests: 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., (Copyright © 2024 Gangaev, van Sleen, Brandhorst, Hoefakker, Prajapati, Singh, Boerma, van der Heiden, Oosting, van der Veldt, Hiltermann, GeurtsvanKessel, Dingemans, Smit, de Vries, Haanen, Kvistborg and van Baarle.)
- Published
- 2024
- Full Text
- View/download PDF
21. Corticosteroids and other immunosuppressants for immune-related adverse events and checkpoint inhibitor effectiveness in melanoma.
- Author
-
Verheijden RJ, Burgers FH, Janssen JC, Putker AE, Veenstra SPGR, Hospers GAP, Aarts MJB, Hehenkamp KW, Doornebosch VLE, Verhaert M, van den Berkmortel FWPJ, Chatzidionysiou K, Llobell A, Barros M, Maria ATJ, Takeji A, García Morillo JS, Lidar M, van Eijs MJM, Blank CU, Aspeslagh S, Piersma D, Kapiteijn E, Labots M, Boers-Sonderen MJ, van der Veldt AAM, Haanen JBAG, May AM, and Suijkerbuijk KPM
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Retrospective Studies, Skin Neoplasms drug therapy, Skin Neoplasms immunology, Skin Neoplasms mortality, Aged, 80 and over, Melanoma drug therapy, Melanoma immunology, Melanoma mortality, Immunosuppressive Agents adverse effects, Immunosuppressive Agents therapeutic use, Immune Checkpoint Inhibitors adverse effects, Adrenal Cortex Hormones therapeutic use, Adrenal Cortex Hormones adverse effects
- Abstract
Background: Recent studies indicate an association between immunosuppression for immune-related adverse events (irAEs) and impaired survival in patients who received immune checkpoint inhibitors. Whether this is related to corticosteroids or second-line immunosuppressants is unknown. In the largest cohort thus far, we assessed the association of immunosuppressant type and dose with survival in melanoma patients with irAEs., Methods: Patients with advanced melanoma who received immunosuppressants for irAEs induced by first-line anti-PD-1 ± anti-CTLA-4 were included from 18 hospitals worldwide. Associations of cumulative and peak dose corticosteroids and use of second-line immunosuppression with survival from start of immunosuppression were assessed using multivariable Cox proportional hazard regression., Results: Among 606 patients, 404 had anti-PD-1 + anti-CTLA-4-related irAEs and 202 had anti-PD-1-related irAEs. 425 patients (70 %) received corticosteroids only; 181 patients (30 %) additionally received second-line immunosuppressants. Median PFS and OS from starting immunosuppression were 4.5 (95 %CI 3.4-8.1) and 31 (95 %CI 15-not reached) months in patients who received second-line immunosuppressants, and 11 (95 %CI 9.4-14) and 55 (95 %CI 41-not reached) months in patients who did not. High corticosteroid peak dose was associated with worse PFS and OS (HR
adj 1.14; 95 %CI 1.01-1.29; HRadj 1.29; 95 %CI 1.12-1.49 for 80vs40mg), while cumulative dose was not. Second-line immunosuppression was associated with worse PFS (HRadj 1.32; 95 %CI 1.02-1.72) and OS (HRadj 1.34; 95 %CI 0.99-1.82) compared with corticosteroids alone., Conclusions: High corticosteroid peak dose and second-line immunosuppressants to treat irAEs are both associated with impaired survival. While immunosuppression is indispensable for treatment of severe irAEs, clinicians should weigh possible detrimental effects on survival against potential disadvantages of undertreatment., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: GAPH reports consultancy/advisory relationships with Amgen, Bristol-Myers Squibb, Roche, Merck Sharp and Dome, Pfizer, Novartis, Sanofi, Pierre Fabre and has received research funding from Bristol-Myers Squibb and Seerave. All paid to institution. MJBA reports consultancy/advisory relationships with Amgen, Bristol Myers Squibb, Novartis, Merck Sharp and Dome, Merck-Pfizer, Pierre Fabre, Sanofi, Astellas and Bayer, and received research funding from Merck-Pfizer. All paid to institution. KC reports consultancy fees from Eli Lilly AbbVie and Pfizer. ATJM has received fees from AbbVie, Actelion, CSL Behring, Experf, Novartis, and Shire and declares speaking fees from AstraZeneca, Sanofi-Aventis and Bristol-Myers Squibb. CUB reports consulting/advisory relationships with AstraZeneca, Bristol-Myers Squibb, GenMab, GSK, Lilly, Merck Sharp and Dome, Novartis, Pfizer, Pierre Fabre, Roche and Third Rock Ventures, and received research funding from 4SC, Bristol-Myers Squibb, NanoString and Novartis. All paid to institution. His is co-founder of and owns shares in Immagene BV and Signature Oncology, and is inventor on several related patents (including submitted): WO 2021/177822 A1, N2027907 and P091040NL2. SA reports consulting/advisory relationships with Merck Sharp and Dome, Sanofi, Roche, Bristol-Myers Squibb, Pfizer, Ipsen and Galapagos. All paid to institution. DP reports consultancy/advisory relationships with Pierre Fabre and Novartis. Partly paid to intstitution. EK reports consultancy/advisory relationships with Bristol Myers Squibb, Novartis, Pierre Fabre, Immunocore and Lilly, and received research grants not related to this paper from Bristol Myers Squibb, Delcath, Novartis and Pierre-Fabre. Not related to current work and paid to institute. ML reports consultancy/advisory relationships with Bristol-Myers Squibb and Janssen-Cilag B.V. All paid to institution. AMMvdV reports consultancy/advisory relationships with Bristol-Myers Squibb, Merck Sharp and Dome, Sanofi, Pfizer, Novartis, Roche, Eisai, Merck, Pierre Fabre and Ipsen. All paid to institution. JBAGH reports consultancy/advisory relationships with Achillus Tx, AstraZenica, BioNTech, Bristol-Myers Squibb, CureVac, GlaxoSmithKline, Imcyse, Iovance Bio, Instil Bio, Ipsen, Merck, Merck Sharp and Dome, Molecular Partners, Neogene TX, Novartis, Pfizer, PokeAcell, Roche, Sanofi, Scenic, T-Knife and TRV, and received research funding from Amgen, Bristol-Myers Squibb, BioNTech, Merck Sharp and Dome, Novartis and Sastra Cell Therapy. All paid to institution. KPMS reports consulting/advisory relationships with Bristol-Myers Squibb, Merck Sharp and Dome, Abbvie, Pierre Fabre, Novartis, Sairopa. She received honoraria from Novartis and Merck Sharp and Dome, and research funding from TigaTx, Bristol Myers Squibb, Philips and Genmab. All paid to institution. All remaining authors have declared no conflicts of interest., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
22. Long-Term Survival in Patients With Advanced Melanoma.
- Author
-
van Not OJ, van den Eertwegh AJM, Jalving H, Bloem M, Haanen JB, van Rijn RS, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, de Groot J W B JW, Hospers GAP, Kapiteijn E, Leeneman B, D P, Stevense-den Boer M, van der Veldt AAM, Vreugdenhil G G, Wouters MWJM, Blokx WAM, and Suijkerbuijk KPM
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Netherlands epidemiology, Ipilimumab therapeutic use, Nivolumab therapeutic use, Skin Neoplasms drug therapy, Skin Neoplasms mortality, Skin Neoplasms pathology, Cohort Studies, Registries, Progression-Free Survival, Prospective Studies, Melanoma drug therapy, Melanoma mortality, Immune Checkpoint Inhibitors therapeutic use
- Abstract
Importance: Long-term survival data from clinical trials show that survival curves of patients with advanced melanoma treated with immune checkpoint inhibitors (ICIs) gradually reach a plateau, suggesting that patients have a chance of achieving long-term survival., Objective: To investigate long-term survival in patients with advanced melanoma treated with ICIs outside clinical trials., Design, Setting, and Participants: Cohort study using prospectively collected data from the nationwide Dutch Melanoma Treatment Registry, including patients in the Netherlands with advanced melanoma treated with first-line ICIs from 2012 to 2019. Data were analyzed from January to September 2023., Exposures: Patients were treated with first-line ipilimumab-nivolumab, antibodies that target programmed cell death (anti-PD-1), or ipilimumab., Main Outcomes and Measures: Progression-free survival (PFS) and melanoma-specific survival were analyzed, and a Cox proportional hazards model was used to investigate factors associated with PFS after reaching partial response (PR) or complete response (CR)., Results: A total of 2490 patients treated with first-line ICIs were included (median [IQR] age, 65.0 [55.3-73.0] years; 1561 male patients [62.7%]). Most patients had an Eastern Cooperative Oncology Group Performance Status of 1 or lower (2202 patients [88.5%]) and normal lactate dehydrogenase levels (1715 patients [68.9%]). PFS for all patients was 23.4% (95% CI, 21.7%-25.2%) after 3 years and 19.7% (95% CI, 18.0%-21.4%) after 5 years. Overall survival for all patients was 44.0% (95% CI, 42.1%-46.1%) after 3 years and 35.9% (95% CI, 33.9%-38.0%) after 5 years. Patients with metastases in 3 or more organ sites had a significantly higher hazard of progression after reaching PR or CR (adjusted hazard ratio, 1.37; 95% CI, 1.11-1.69)., Conclusions and Relevance: This cohort study of patients with advanced melanoma treated with ICIs in clinical practice showed that their survival reached a plateau, comparable with patients participating in clinical trials. These findings can be used in daily clinical practice to guide long-term surveillance strategies and inform both physicians and patients regarding long-term treatment outcomes.
- Published
- 2024
- Full Text
- View/download PDF
23. Detection of Drug-induced Interstitial Lung Disease Caused by Cancer Treatment Using Electronic Nose Exhaled Breath Analysis.
- Author
-
van der Sar IG, Wijsenbeek MS, Dumoulin DW, Jager A, van der Veldt AAM, Rossius MJP, Dingemans AC, and Moor CC
- Subjects
- Humans, Antineoplastic Agents adverse effects, Exhalation, Neoplasms drug therapy, Breath Tests methods, Electronic Nose, Lung Diseases, Interstitial diagnosis, Lung Diseases, Interstitial chemically induced
- Published
- 2024
- Full Text
- View/download PDF
24. From decision to reflection: understanding the experiences and unmet care needs of patients treated with immunotherapy for melanoma in the adjuvant or metastatic setting.
- Author
-
Kamminga NCW, van der Veldt AAM, Wakkee M, van den Berge FR, van der Beek LAA, Joosen MCW, Joosse A, de Joode K, Nijsten TEC, and Lugtenberg M
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Adult, Decision Making, Focus Groups, Neoplasm Metastasis, Qualitative Research, Aged, 80 and over, Melanoma therapy, Melanoma drug therapy, Melanoma immunology, Immune Checkpoint Inhibitors therapeutic use, Immunotherapy methods
- Abstract
Background: Despite increased use of immune checkpoint inhibitors (ICIs) in patients with advanced melanoma, little is known about patient experiences during this treatment. This study aimed to gain an in-depth understanding of experiences and unmet care needs of patients treated in the adjuvant or metastatic setting for advanced melanoma regarding their ICI treatment trajectory., Methods: Interviews and focus groups were conducted among 35 patients treated with ICIs in the adjuvant setting for completely resected stage III (n = 14), or in the metastatic setting for irresectable stage IV (n = 21) melanoma. A thorough thematic content analysis was conducted., Results: Three main themes were identified. When (1) dealing with uncertainty in the decision-making process, adjuvant patients explored the pros and cons, whereas metastatic patients considered immunotherapy their only viable option. Both groups expressed the need for additional guidance. In (2) navigating the immunotherapy course, both perceived the trajectory as intense, experienced a major impact on their and their (close) relatives' lives, and felt the need to (re)gain control. When (3) looking back on the immunotherapy experience, metastatic patients generally felt relieved, while among adjuvant patients, feelings of doubt regarding their choice for ICIs were also reported., Conclusions: ICI treatment is perceived as intensive for both patient groups, facing both comparable and distinct challenges throughout the treatment trajectory, underscoring the need for stage-specific, individualised guidance. Options regarding flexible follow-ups, low-threshold contact and psychosocial support throughout the treatment trajectory should be explored., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
25. Safe Stop IPI-NIVO trial: early discontinuation of nivolumab upon achieving a complete or partial response in patients with irresectable stage III or metastatic melanoma treated with first-line ipilimumab-nivolumab - study protocol.
- Author
-
Janssen JC, van Dijk B, de Joode K, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, van den Eertwegh AJM, de Groot JWB, Jalving M, de Jonge MJA, Joosse A, Kapiteijn E, Kamphuis-Huismans AM, Naipal KAT, Piersma D, Rikhof B, Westgeest HM, Vreugdenhil G, Oomen-de Hoop E, Mulder EEAP, and van der Veldt AAM
- Subjects
- Female, Humans, Male, Immune Checkpoint Inhibitors adverse effects, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors administration & dosage, Neoplasm Staging, Netherlands, Prospective Studies, Skin Neoplasms drug therapy, Skin Neoplasms pathology, Withholding Treatment, Multicenter Studies as Topic, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Ipilimumab administration & dosage, Ipilimumab adverse effects, Ipilimumab therapeutic use, Melanoma drug therapy, Melanoma pathology, Nivolumab administration & dosage, Nivolumab adverse effects, Nivolumab therapeutic use
- Abstract
Background: Patients with irresectable stage III or metastatic melanoma presenting with poor prognostic factors are usually treated with a combination of immune checkpoint inhibitors (ICIs), consisting of ipilimumab and nivolumab. This combination therapy is associated with severe immune related adverse events (irAEs) in about 60% of patients. In current clinical practice, patients are usually treated with ICIs for up to two years or until disease progression or the occurrence of unacceptable AEs. The incidence of irAEs gradually increases with duration of treatment. While durable tumour responses have been observed after early discontinuation of treatment, no consensus has been reached on optimal treatment duration. The objective of the Safe Stop IPI-NIVO trial is to evaluate whether early discontinuation of ICIs is safe in patients with irresectable stage III or metastatic melanoma who are treated with combination therapy., Methods: The Safe Stop IPI-NIVO trial is a nationwide, multicentre, prospective, single-arm, interventional study in the Netherlands. A total of 80 patients with irresectable stage III or metastatic melanoma who are treated with combination therapy of ipilimumab-nivolumab and have a complete or partial response (CR/PR) according to RECIST v1.1 will be included to early discontinue maintenance therapy with anti-PD-1. The primary endpoint is the rate of ongoing response at 12 months after start of ICI. Secondary endpoints include ongoing response at 24 months, disease control at different time points, melanoma specific and overall survival, the incidence of irAEs and health-related quality of life., Discussion: From a medical, healthcare and economic perspective, overtreatment should be prevented and shorter treatment duration of ICIs is preferred. If early discontinuation of ICIs is safe for patients who are treated with the combination of ipilimumab-nivolumab, the treatment duration of nivolumab could be shortened in patients with a favourable tumour response., Trial Registration: ClinicalTrials.gov ID NCT05652673, registration date: 08-12-2022., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
26. Multi-omic analysis identifies hypoalbuminemia as independent biomarker of poor outcome upon PD-1 blockade in metastatic melanoma.
- Author
-
Leek LVM, Notohardjo JCL, de Joode K, Velker EL, Haanen JBAG, Suijkerbuijk KPM, Aarts MJB, de Groot JWB, Kapiteijn E, van den Berkmortel FWPJ, Westgeest HM, de Gruijl TD, Retel VP, Cuppen E, van der Veldt AAM, Labots M, Voest EE, van de Haar J, and van den Eertwegh AJM
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Prognosis, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor metabolism, Adult, Neoplasm Metastasis, L-Lactate Dehydrogenase blood, L-Lactate Dehydrogenase metabolism, Aged, 80 and over, Multiomics, Melanoma drug therapy, Melanoma pathology, Melanoma metabolism, Hypoalbuminemia, Biomarkers, Tumor blood, Immune Checkpoint Inhibitors therapeutic use
- Abstract
We evaluated the prognostic value of hypoalbuminemia in context of various biomarkers at baseline, including clinical, genomic, transcriptomic, and blood-based markers, in patients with metastatic melanoma treated with anti-PD-1 monotherapy or anti-PD-1/anti-CTLA-4 combination therapy (n = 178). An independent validation cohort (n = 79) was used to validate the performance of hypoalbuminemia compared to serum LDH (lactate dehydrogenase) levels. Pre-treatment hypoalbuminemia emerged as the strongest predictor of poor outcome for both OS (HR = 4.01, 95% CI 2.10-7.67, Cox P = 2.63e-05) and PFS (HR = 3.72, 95% CI 2.06-6.73, Cox P = 1.38e-05) in univariate analysis. In multivariate analysis, the association of hypoalbuminemia with PFS was independent of serum LDH, IFN-γ signature expression, TMB, age, ECOG PS, treatment line, treatment type (combination or monotherapy), brain and liver metastasis (HR = 2.76, 95% CI 1.24-6.13, Cox P = 0.0131). Our validation cohort confirmed the prognostic power of hypoalbuminemia for OS (HR = 1.98, 95% CI 1.16-3.38; Cox P = 0.0127) and was complementary to serum LDH in analyses for both OS (LDH-adjusted HR = 2.12, 95% CI 1.2-3.72, Cox P = 0.00925) and PFS (LDH-adjusted HR = 1.91, 95% CI 1.08-3.38, Cox P = 0.0261). In conclusion, pretreatment hypoalbuminemia was a powerful predictor of outcome in ICI in melanoma and showed remarkable complementarity to previously established biomarkers, including high LDH., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
27. A prediction model for response to immune checkpoint inhibition in advanced melanoma.
- Author
-
van Duin IAJ, Verheijden RJ, van Diest PJ, Blokx WAM, El-Sharouni MA, Verhoeff JJC, Leiner T, van den Eertwegh AJM, de Groot JWB, van Not OJ, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Haanen JBAG, Hospers GAP, Piersma D, van Rijn RS, van der Veldt AAM, Vreugdenhil G, Wouters MWJM, Stevense-den Boer MAM, Boers-Sonderen MJ, Kapiteijn E, Suijkerbuijk KPM, and Elias SG
- Subjects
- Humans, Immune Checkpoint Inhibitors therapeutic use, Ipilimumab therapeutic use, Nivolumab therapeutic use, Retrospective Studies, Melanoma pathology, Skin Neoplasms pathology
- Abstract
Predicting who will benefit from treatment with immune checkpoint inhibition (ICI) in patients with advanced melanoma is challenging. We developed a multivariable prediction model for response to ICI, using routinely available clinical data including primary melanoma characteristics. We used a population-based cohort of 3525 patients with advanced cutaneous melanoma treated with anti-PD-1-based therapy. Our prediction model for predicting response within 6 months after ICI initiation was internally validated with bootstrap resampling. Performance evaluation included calibration, discrimination and internal-external cross-validation. Included patients received anti-PD-1 monotherapy (n = 2366) or ipilimumab plus nivolumab (n = 1159) in any treatment line. The model included serum lactate dehydrogenase, World Health Organization performance score, type and line of ICI, disease stage and time to first distant recurrence-all at start of ICI-, and location and type of primary melanoma, the presence of satellites and/or in-transit metastases at primary diagnosis and sex. The over-optimism adjusted area under the receiver operating characteristic was 0.66 (95% CI: 0.64-0.66). The range of predicted response probabilities was 7%-81%. Based on these probabilities, patients were categorized into quartiles. Compared to the lowest response quartile, patients in the highest quartile had a significantly longer median progression-free survival (20.0 vs 2.8 months; P < .001) and median overall survival (62.0 vs 8.0 months; P < .001). Our prediction model, based on routinely available clinical variables and primary melanoma characteristics, predicts response to ICI in patients with advanced melanoma and discriminates well between treated patients with a very good and very poor prognosis., (© 2024 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2024
- Full Text
- View/download PDF
28. BRAF/MEK inhibitor rechallenge in advanced melanoma patients.
- Author
-
Van Not OJ, van den Eertwegh AJM, Haanen JB, van Rijn RS, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, de Groot JWWB, Hospers GAP, Kapiteijn E, Bloem M, Piersma D, Stevense-den Boer M, Verheijden RJ, van der Veldt AAM, Wouters MWJM, Blokx WAM, and Suijkerbuijk KPM
- Subjects
- Humans, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mutation, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Retrospective Studies, Brain Neoplasms etiology, Brain Neoplasms pathology, Melanoma drug therapy, Melanoma pathology
- Abstract
Background: Effectivity of BRAF(/MEK) inhibitor rechallenge has been described in prior studies. However, structured data are largely lacking., Methods: Data from all advanced melanoma patients treated with BRAFi(/MEKi) rechallenge were retrieved from the Dutch Melanoma Treatment Registry. The authors analyzed objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) for both first treatment and rechallenge. They performed a multivariable logistic regression and a multivariable Cox proportional hazards model to assess factors associated with response and survival., Results: The authors included 468 patients in the largest cohort to date who underwent at least two treatment episodes of BRAFi(/MEKi). Following rechallenge, ORR was 43%, median PFS was 4.6 months (95% confidence interval [CI], 4.1-5.2), and median OS was 8.2 months (95% CI, 7.2-9.4). Median PFS after rechallenge for patients who discontinued first BRAFi(/MEKi) treatment due to progression was 3.1 months (95% CI, 2.7-4.0) versus 5.2 months (95% CI, 4.5-5.9) for patients who discontinued treatment for other reasons. Discontinuing first treatment due to progression and lactate dehydrogenase (LDH) levels greater than two times the upper limit of normal were associated with lower odds of response and worse PFS and OS. Symptomatic brain metastases were associated with worse survival, whereas a longer treatment interval between first treatment and rechallenge was associated with better survival. Responding to the first BRAFi(/MEKi) treatment was not associated with response or survival., Conclusions: This study confirms that patients benefit from rechallenge. Elevated LDH levels, symptomatic brain metastases, and discontinuing first BRAFi(/MEKi) treatment due to progression are associated with less benefit from rechallenge. A prolonged treatment interval is associated with more benefit from rechallenge., (© 2024 The Authors. Cancer published by Wiley Periodicals LLC on behalf of American Cancer Society.)
- Published
- 2024
- Full Text
- View/download PDF
29. Whole-genome mapping of APOBEC mutagenesis in metastatic urothelial carcinoma identifies driver hotspot mutations and a novel mutational signature.
- Author
-
Nakauma-González JA, Rijnders M, Noordsij MTW, Martens JWM, van der Veldt AAM, Lolkema MPJ, Boormans JL, and van de Werken HJG
- Subjects
- Humans, Mutagenesis genetics, Mutation genetics, Chromosome Mapping, Carcinoma, Transitional Cell genetics, Urinary Bladder Neoplasms genetics
- Abstract
Apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC) enzymes mutate specific DNA sequences and hairpin-loop structures, challenging the distinction between passenger and driver hotspot mutations. Here, we characterized 115 whole genomes of metastatic urothelial carcinoma (mUC) to identify APOBEC mutagenic hotspot drivers. APOBEC-associated mutations were detected in 92% of mUCs and were equally distributed across the genome, while APOBEC hotspot mutations (ApoHMs) were enriched in open chromatin. Hairpin loops were frequent targets of didymi (twins in Greek), two hotspot mutations characterized by the APOBEC SBS2 signature, in conjunction with an uncharacterized mutational context (Ap[C>T]). Next, we developed a statistical framework that identified ApoHMs as drivers in coding and non-coding genomic regions of mUCs. Our results and statistical framework were validated in independent cohorts of 23 non-metastatic UCs and 3,744 samples of 17 metastatic cancers, identifying cancer-type-specific drivers. Our study highlights the role of APOBEC in cancer development and may contribute to developing novel targeted therapy options for APOBEC-driven cancers., Competing Interests: Declaration of interests J.L.B. has received research support from Merck AG/Pfizer, Janssen, and Merck Sharp & Dohme and consultancy fees from Merck Sharp & Dohme, Bristol-Myers Squibb, Astellas, AstraZeneca, Ipsen, and Janssen (all paid to the Erasmus MC Cancer Institute). M.P.J.L. has received research support from JnJ, Sanofi, Astellas, and MSD and consultancy fees from Incyte, Amgen, JnJ, Bayer, Servier, Roche, INCa, Pfizer, Sanofi, Astellas, AstraZeneca, Merck Sharp & Dohme, Novartis, Julius Clinical, and the Hartwig Medical Foundation (all paid to the Erasmus MC Cancer Institute). J.W.M.M. has received research support from Pfizer, Sanofi, GSK, Therawis Cergentis, and Philips (all paid to the Erasmus MC Cancer Institute) and one consultancy fee from Novartis. A.A.M.v.d.V. has received consultancy fees from BMS, MSD, Pfizer, Novartis, Eisai, Sanofi, Pierre Fabre, Ipsen, and Roche (all paid to the Erasmus MC Cancer Institute)., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
30. Development and validation of a novel model to predict recurrence-free survival and melanoma-specific survival after sentinel lymph node biopsy in patients with melanoma: an international, retrospective, multicentre analysis.
- Author
-
Stassen RC, Maas CCHM, van der Veldt AAM, Lo SN, Saw RPM, Varey AHR, Scolyer RA, Long GV, Thompson JF, Rutkowski P, Keilholz U, van Akkooi ACJ, Verhoef C, van Klaveren D, and Grünhagen DJ
- Subjects
- Humans, Male, Female, Sentinel Lymph Node Biopsy, Retrospective Studies, Lymphatic Metastasis, Prognosis, Melanoma pathology, Skin Neoplasms pathology, Sentinel Lymph Node surgery, Sentinel Lymph Node pathology, Lymphadenopathy pathology
- Abstract
Background: The introduction of adjuvant systemic treatment for patients with high-risk melanomas necessitates accurate staging of disease. However, inconsistencies in outcomes exist between disease stages as defined by the American Joint Committee on Cancer (8th edition). We aimed to develop a tool to predict patient-specific outcomes in people with melanoma rather than grouping patients according to disease stage., Methods: Patients older than 13 years with confirmed primary melanoma who underwent sentinel lymph node biopsy (SLNB) between Oct 29, 1997, and Nov 11, 2013, at four European melanoma centres (based in Berlin, Germany; Amsterdam and Rotterdam, the Netherlands; and Warsaw, Poland) were included in the development cohort. Potential predictors of recurrence-free and melanoma-specific survival assessed were sex, age, presence of ulceration, primary tumour location, histological subtype, Breslow thickness, sentinel node status, number of sentinel nodes removed, maximum diameter of the largest sentinel node metastasis, and Dewar classification. A prognostic model and nomogram were developed to predict 5-year recurrence-free survival on a continuous scale in patients with stage pT1b or higher melanomas. This model was also calibrated to predict melanoma-specific survival. Model performance was assessed by discrimination (area under the time-dependent receiver operating characteristics curve [AUC]) and calibration. External validation was done in a cohort of patients with primary melanomas who underwent SLNB between Jan 30, 1997, and Dec 12, 2013, at the Melanoma Institute Australia (Sydney, NSW, Australia)., Findings: The development cohort consisted of 4071 patients, of whom 2075 (51%) were female and 1996 (49%) were male. 889 (22%) had sentinel node-positive disease and 3182 (78%) had sentinel node-negative disease. The validation cohort comprised 4822 patients, of whom 1965 (41%) were female and 2857 (59%) were male. 891 (18%) had sentinel node-positive disease and 3931 (82%) had sentinel node-negative disease. Median follow-up was 4·8 years (IQR 2·3-7·8) in the development cohort and 5·0 years (2·2-8·9) in the validation cohort. In the development cohort, 5-year recurrence-free survival was 73·5% (95% CI 72·0-75·1) and 5-year melanoma-specific survival was 86·5% (85·3-87·8). In the validation cohort, the corresponding estimates were 66·1% (64·6-67·7) and 83·3% (82·0-84·6), respectively. The final model contained six prognostic factors: sentinel node status, Breslow thickness, presence of ulceration, age at SLNB, primary tumour location, and maximum diameter of the largest sentinel node metastasis. In the development cohort, for the model's prediction of recurrence-free survival, the AUC was 0·80 (95% CI 0·78-0·81); for prediction of melanoma-specific survival, the AUC was 0·81 (0·79-0·84). External validation showed good calibration for both outcomes, with AUCs of 0·73 (0·71-0·75) and 0·76 (0·74-0·78), respectively., Interpretation: Our prediction model and nomogram accurately predicted patient-specific risk probabilities for 5-year recurrence-free and melanoma-specific survival. These tools could have important implications for clinical decision making when considering adjuvant treatments in patients with high-risk melanomas., Funding: Erasmus Medical Centre Cancer Institute., Competing Interests: Declaration of interests RCS reports honoraria (paid to their institution) from Amgen. AAMvdV reports consultancy fees (paid to their institution) from Bristol Myers Squibb, Merck Sharp & Dohme, Sanofi, Pierre Fabre, Ipsen, Eisai, Merck, Pfizer, Novartis, and Roche. RPMS has received honoraria for advisory board participation from Merck Sharp & Dohme, Novartis, and Qbiotics, and speaker fees from Bristol Myers Squibb and Novartis. AHRV has received honoraria from Novartis. RAS has received fees for professional services from MetaOptima Technology, F Hoffmann-La Roche, Evaxion, Provectus, Qbiotics, Novartis, Merck Sharp & Dohme, NeraCare, Amgen, Bristol Myers Squibb, Myriad Genetics, and GlaxoSmithKline. GVL reports consultancy or advisory fees from Agenus, Amgen, Array Biopharma, AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Evaxion, Hexal, Highlight Therapeutics, Innovent Biologics, Novartis, OncoSec, PHMR, Pierre Fabre, Provectus, Qbiotics, and Regeneron. JFT has received honoraria for advisory board participation from Bristol Myers Squibb, Merck Sharp & Dohme, GlaxoSmithKline, and Provectus, and travel and conference support from GlaxoSmithKline, Provectus, and Novartis. PR reports research funding (paid to their institution) from Merck Sharp & Dohme and Pfizer, and has received honoraria for lectures and advisory board participation from Merck Sharp & Dohme, Bristol Myers Squibb, Novartis, Pierre Fabre, Sanofi, Merck, Philogen, and Blueprint Medicines. UK has received consulting fees from Merck Sharp & Dohme, travel support from Merck Serono and Pfizer, and grants for educational activities from Merck Serono, Bristol Myers Squibb, and Pierre-Fabre. ACJvA reports honoraria for consultancy and serving on advisory boards (all paid to their institution) from Amgen, Bristol Myers Squibb, Merck Sharp & Dohme, Merck, Pfizer, Neracare, Novartis, Pierre Fabre, Provectus, Sanofi, Sirius Medical, and 4SC, and research grants (paid to their institution) from Amgen, Merck, and Pfizer. DvK has participated on the data and safety monitoring boards of the FAST CABG and Multivessel Talent trials. DJG has served on data and safety monitoring boards for Amgen and Novartis (funds paid to their institution). All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
31. Cemiplimab in locally advanced or metastatic cutaneous squamous cell carcinoma: prospective real-world data from the DRUG Access Protocol.
- Author
-
Verkerk K, Geurts BS, Zeverijn LJ, van der Noort V, Verheul HMW, Haanen JBAG, van der Veldt AAM, Eskens FALM, Aarts MJB, van Herpen CML, Jalving M, Gietema JA, Devriese LA, Labots M, Barjesteh van Waalwijk van Doorn-Khosrovani S, Smit EF, and Bloemendal HJ
- Abstract
Background: The DRUG Access Protocol provides patients with cancer access to registered anti-cancer drugs that are awaiting reimbursement in the Netherlands and simultaneously collects prospective real-world data (RWD). Here, we present RWD from PD-1 blocker cemiplimab in patients with locally advanced or metastatic cutaneous squamous cell carcinoma (laCSCC; mCSCC)., Methods: Patients with laCSCC or mCSCC received cemiplimab 350 mg fixed dose every three weeks. Primary endpoints were objective clinical benefit rate (CBR), defined as objective response (OR) or stable disease (SD) at 16 weeks, physician-assessed CBR, defined as clinician's documentation of improved disease or SD based on evaluation of all available clinical parameters at 16 weeks, objective response rate (ORR), and safety, defined as grade ≥ 3 treatment related adverse events (TRAEs) occurring up to 30 days after last drug administration. Secondary endpoints included duration of response (DoR), progression-free survival (PFS), and overall survival (OS)., Findings: Between February 2021 and December 2022, 151 patients started treatment. Objective and physician-assessed CBR were 54.3% (95% CI, 46.0-62.4) and 59.6% (95% CI, 51.3-67.5), respectively. ORR was 35.1% (95% CI, 27.5-43.3). After a median follow-up of 15.2 months, median DoR was not reached. Median PFS and OS were 12.2 (95% CI, 7.0-not reached) and 24.2 months (95% CI, 18.8-not reached), respectively. Sixty-eight TRAEs occurred in 29.8% of patients. Most commonly reported TRAE was a kidney transplant rejection (9.5%)., Interpretation: Cemiplimab proved highly effective and safe in this real-world cohort of patients with laCSCC or mCSCC, confirming its therapeutic value in the treatment of advanced CSCC in daily clinical practice., Funding: The DRUG Access Protocol is supported by all participating pharmaceutical companies: Bayer, Janssen, Lilly, Merck, Novartis, Roche, and Sanofi., Competing Interests: John B.A.G. Haanen reports research grants to the institution from Asher Bio, Amgen, BioNTech, Bristol Myers Squibb, Novartis, and Sastra Cell Therapy, consultation fees to the institution from Achilles Tx, AstraZeneca, BioNTech, Bristol Myers Squibb, CureVac, Eisai, GlaxoSmithKline, Imcyse, Immunocore, Instil Bio, Iovance Bio, Merck, Merck Sharp & Dohme, Neogene Tx, Novartis, Obsidian Tx, Roche, Sanofi, Sastra Cell Therapy, Third Rock Ventures, and T-Knife, and stock options from Neogene Tx and Sastra Cell Therapy. Mathilde Jalving received compensation to the institution for advisory roles for Pierre Fabre, Merck, and Novartis. Lot A. Devriese received compensation to the institution for advisory roles and education for Bristol Myers Squibb, Merck Sharp & Dohme, and Incyte. Astrid A.M. van der Veldt received compensation to the institution for advisory roles for Bristol Myers Squibb, Merck Sharp & Dohme, Eisai, Ipsen, Novartis, Pierre Fabre, Pfizer, Roche, and Sanofi. Maureen J.B. Aarts reports research grants to the institution from Merck-Pfizer and consultation fees to the institution from being on the advisory board from Amgen, Bristol Myers Squibb, Novartis, Merck Sharp & Dohme, Merck-Pfizer, Pierre Fabre, Sanofi, Astellas, and Bayer. Carla M.L. van Herpen reports research grants to the institution from Astra Zeneca, Bayer, Bristol Myers Squibb, Elevar, Ipsen, Merck Sharp & Dohme, Merck, Novartis, and Sanofi, payments for lectures to the institution from ICCR, NIFU, RIVM, Roije Congressen, Nederlands Paramedisch Instituut, Elevar, and Uitgeverij Jaap, and payments to the institution for participating on an advisory board to the institution from Elevar. Mariette Labots received compensation to the institution for education for Bristol Myers Squibb and Janssen. Sahar Barjesteh van Waalwijk van Doorn-Khosrovani reports the PRIME-ROSE—A European precision cancer medicine trial network and implementation initiative funded by the EU Cancer Mission; grant no. 101104269 paid to the Leiden University Medical Center and travel expenses for attending meetings at the Nordic Precision Medicine Forum and Center for Innovation in Regulatory Science. Egbert F. Smit reports compensation to the institution for advisory roles at AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Daiichi Sankyo, Eli Lilly, Janssen, Merck Sharp & Dohme, Roche, Sanofi, and Takeda, personal compensation for education from Boehringer Ingelheim and Daiichi Sankyo, and compensation to the institution for participation in the data safety monitoring board of DSI. The other authors have no conflicts of interests to disclose., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
32. Correlation between Histopathological Prognostic Tumor Characteristics and [ 18 F]FDG Uptake in Corresponding Metastases in Newly Diagnosed Metastatic Breast Cancer.
- Author
-
Boers J, Eisses B, Zwager MC, van Geel JJL, Bensch F, de Vries EFJ, Hospers GAP, Glaudemans AWJM, Brouwers AH, den Dekker MAM, Elias SG, Kuip EJM, van Herpen CML, Jager A, van der Veldt AAM, Oprea-Lager DE, de Vries EGE, van der Vegt B, Menke-van der Houven van Oordt WC, and Schröder CP
- Abstract
Background: In metastatic breast cancer (MBC), [
18 F]fluorodeoxyglucose positron emission tomography/computed tomography ([18 F]FDG-PET/CT) can be used for staging. We evaluated the correlation between BC histopathological characteristics and [18 F]FDG uptake in corresponding metastases., Patients and Methods: Patients with non-rapidly progressive MBC of all subtypes prospectively underwent a baseline histological metastasis biopsy and [18 F]FDG-PET. Biopsies were assessed for estrogen, progesterone, and human epidermal growth factor receptor 2 (ER, PR, HER2); Ki-67; and histological subtype. [18 F]FDG uptake was expressed as maximum standardized uptake value (SUVmax ) and results were expressed as geometric means., Results: Of 200 patients, 188 had evaluable metastasis biopsies, and 182 of these contained tumor. HER2 positivity and Ki-67 ≥ 20% were correlated with higher [18 F]FDG uptake (estimated geometric mean SUVmax 10.0 and 8.8, respectively; p = 0.0064 and p = 0.014). [18 F]FDG uptake was lowest in ER-positive/HER2-negative BC and highest in HER2-positive BC (geometric mean SUVmax 6.8 and 10.0, respectively; p = 0.0058). Although [18 F]FDG uptake was lower in invasive lobular carcinoma ( n = 31) than invasive carcinoma NST ( n = 146) (estimated geometric mean SUVmax 5.8 versus 7.8; p = 0.014), the metastasis detection rate was similar., Conclusions: [18 F]FDG-PET is a powerful tool to detect metastases, including invasive lobular carcinoma. Although BC histopathological characteristics are related to [18 F]FDG uptake, [18 F]FDG-PET and biopsy remain complementary in MBC staging (NCT01957332).- Published
- 2024
- Full Text
- View/download PDF
33. Gene-expression-based T-Cell-to-Stroma Enrichment (TSE) score predicts response to immune checkpoint inhibitors in urothelial cancer.
- Author
-
Rijnders M, Nakauma-González JA, Robbrecht DGJ, Gil-Jimenez A, Balcioglu HE, Oostvogels AAM, Aarts MJB, Boormans JL, Hamberg P, van der Heijden MS, Szabados BE, van Leenders GJLH, Mehra N, Voortman J, Westgeest HM, de Wit R, van der Veldt AAM, Debets R, and Lolkema MP
- Subjects
- Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Prospective Studies, T-Lymphocytes, B7-H1 Antigen, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell genetics, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms genetics
- Abstract
Immune checkpoint inhibitors (ICI) improve overall survival in patients with metastatic urothelial cancer (mUC), but therapeutic success at the individual patient level varies significantly. Here we identify predictive markers of response, based on whole-genome DNA (n = 70) and RNA-sequencing (n = 41) of fresh metastatic biopsy samples, collected prior to treatment with pembrolizumab. We find that PD-L1 combined positivity score does not, whereas tumor mutational burden and APOBEC mutagenesis modestly predict response. In contrast, T cell-to-stroma enrichment (TSE) score, computed from gene expression signature data to capture the relative abundance of T cells and stromal cells, predicts response to immunotherapy with high accuracy. Patients with a positive and negative TSE score show progression free survival rates at 6 months of 67 and 0%, respectively. The abundance of T cells and stromal cells, as reflected by the TSE score is confirmed by immunofluorescence in tumor tissue, and its good performance in two independent ICI-treated cohorts of patients with mUC (IMvigor210) and muscle-invasive UC (ABACUS) validate the predictive power of the TSE score. In conclusion, the TSE score represents a clinically applicable metric that potentially supports the prospective selection of patients with mUC for ICI treatment., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
34. Improving survival in advanced melanoma patients: a trend analysis from 2013 to 2021.
- Author
-
van Not OJ, van den Eertwegh AJM, Haanen JB, Blank CU, Aarts MJB, van Breeschoten J, van den Berkmortel FWPJ, de Groot JB, Hospers GAP, Ismail RK, Kapiteijn E, Bloem M, De Meza MM, Piersma D, van Rijn RS, Stevense-den Boer MAM, van der Veldt AAM, Vreugdenhil G, Boers-Sonderen MJ, Blokx WAM, Wouters MWJM, and Suijkerbuijk KPM
- Abstract
Background: The prognosis of advanced melanoma patients has significantly improved over the years. We aimed to evaluate the survival per year of diagnosis., Methods: All systemically treated patients diagnosed with advanced melanoma from 2013 to 2021 were included from the Dutch Melanoma Treatment Registry. Baseline characteristics and overall survival (OS) were compared between the different years of diagnosis. A multivariable Cox proportional hazards model was used to estimate the association between year of diagnosis and OS., Findings: For this cohort study, we included 6260 systemically treated advanced melanoma patients. At baseline, there was an increase over the years in age, the percentage of patients with an ECOG PS ≥ 2, with brain metastases, and a synchronous diagnosis of primary and unresectable melanoma. Median OS increased from 11.2 months (95% CI 10.0-12.4) for patients diagnosed in 2013 to 32.0 months (95% CI 26.6-36.7) for patients diagnosed in 2019. Median OS was remarkably lower for patients diagnosed in 2020 (26.6 months; 95% CI 23.9-35.1) and 2021 (24.0 months; 95% CI 20.4-NR). Patients diagnosed in 2020 and 2021 had a higher hazard of death compared to patients diagnosed in 2019, although this was not significant. The multivariable Cox regression showed a lower hazard of death for the years of diagnosis after 2013. In contrast, patients diagnosed in 2020 and 2021 had a higher hazard of death compared to patients diagnosed in 2019., Interpretation: After a continuous survival improvement for advanced melanoma patients between 2013 and 2019, outcomes of patients diagnosed in 2020 and 2021 seem poorer. This trend of decreased survival remained after correcting for known prognostic factors and previous neoadjuvant or adjuvant treatment, suggesting that it is explained by unmeasured factors, which-considering the timing-could be COVID-19-related., Funding: For the Dutch Melanoma Treatment Registry (DMTR), the Dutch Institute for Clinical Auditing foundation received a start-up grant from governmental organization The Netherlands Organization for Health Research and Development (ZonMW, project number 836002002). The DMTR is structurally funded by Bristol-Myers Squibb, Merck Sharpe & Dohme, Novartis, and Roche Pharma. Roche Pharma stopped funding in 2019, and Pierre Fabre started funding the DMTR in 2019. For this work, no funding was granted., Competing Interests: AvdE has advisory relationships with Amgen, Bristol Myers Squibb, Roche, Novartis, MSD, Pierre Fabre, Sanofi, Pfizer, Ipsen, Merck and has received research study grants not related to this paper from Sanofi, Bristol Myers Squibb, Idera and TEVA and has received travel expenses from MSD Oncology, Roche, Pfizer, Pierre Fabre, and Sanofi. JH has advisory relationships with AstraZeneca, Achilles Therapeutics, BioNTech, BMS, CureVac, Iovance Bio, Eisai, Instil Bio, Imcyse, MSD, Neogene Therapeutics, Novartis, Roche, Sanofi, Sastra Cell Therapy, TRV, and T-Knife. And has received research grants not related to this paper from Asher Bio, Amgen, Bristol Myers Squibb, BioNTech, Novartis, and Sastra Cell Therapy. All grants were paid to the institutions. MA has advisory board/consultancy honoraria from Amgen, Bristol Myers Squibb, Novartis, MSD-Merck, Merck-Pfizer, Pierre Fabre, Sanofi, Astellas, Bayer. She received travel expenses from Astella, BMS, Janssen, Pfizer, and Sanofi and a research grant from Merck-Pfizer. Not related to current work and paid to institute. GH consultancy/advisory relationships with Amgen, Bristol Myers Squibb, Roche, MSD, Pfizer, Novartis, Sanofi, Pierre Fabre and has received research grants not related to this paper from Bristol Myers Squibb and Seerave. All payments were paid to the institution. RI has no declarations of interest for this research, but is employed at MSD since January 2022. DP has advisory relationships/consultancy honororia with Pierre Fabre and Novartis and received travel expenses from Pierre Fabre. AvdV has consultancy relationships with Bristol Myers Squibb, MSD, Roche, Novartis, Pierre Fabre, Pfizer, Sanofi, Ipsen, Eisai all paid to the institute. KS has consulting/advisory relationship with Abbvie, Pierre Fabre, and Sairopa. She received honoraria received from Novartis, Roche, Merck Sharp and Dome and research funding not related to this paper from Genmab, TigaTx, Bristol Myers Squibb, and Philips. All paid to institution. All remaining authors have declared no conflicts of interest., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
35. The Predictive Value of FDG PET/CT for Determining Progression-Free Survival in Advanced Stage III-IV BRAF -Mutated Melanoma Patients Treated With Targeted Therapy-What Can Be Learned From Progression?
- Author
-
van der Hiel B, Aalbersberg EA, van den Eertwegh AJM, de Wit-van der Veen LJ, Stokkel MPM, Lopez-Yurda M, Boellaard R, Kapiteijn EW, Hospers GAP, Aarts MJB, de Vos FYFL, Boers-Sonderen MJ, van der Veldt AAM, de Groot JWB, and Haanen JBAG
- Subjects
- Humans, Positron Emission Tomography Computed Tomography, Fluorodeoxyglucose F18, Proto-Oncogene Proteins B-raf genetics, Progression-Free Survival, Prospective Studies, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Melanoma diagnostic imaging, Melanoma drug therapy, Melanoma genetics, Skin Neoplasms pathology
- Abstract
Purpose: The aims of this study were to investigate whether (early) PERCIST response monitoring with 18 F-FDG PET/CT is predictive for progression-free survival (PFS) in unresectable stage III or IV melanoma patients treated with BRAF/MEK inhibitor (MEKi) and to define dissemination patterns at progression with a lesion-based evaluation in direct comparison to baseline to improve our understanding of 18 F-FDG PET/CT during BRAF/MEKi., Patients and Methods: This prospective multicenter single-arm study included 70 patients with unresectable stage III/IV BRAF -mutated melanoma who underwent contrast-enhanced CT and 18 F-FDG PET/CT at baseline and 2 and 7 weeks during treatment with vemurafenib plus cobimetinib and at progression if possible. Tumor response assessment was done with RECIST1.1 and PERCIST. Follow-up PET/CT scans were visually compared with baseline to assess dissemination patterns., Results: Using RECIST1.1, PFS was not significantly different between the response groups ( P = 0.26). At 2 weeks, PERCIST median PFS was 15.7 months for patients with complete metabolic response (CMR) versus 8.3 months for non-CMR ( P = 0.035). The hazards ratio (HR) for progression/death in non-CMR versus CMR was 1.99 (95% confidence interval [CI], 1.03-3.84; P = 0.040) and 1.77 (95% CI, 0.91-3.43; P = 0.0935) when adjusting for lactate dehydrogenase (LDH). At 7 weeks, median PFS for PERCIST CMR was 16.7 months versus 8.5 months for non-CMR ( P = 0.0003). The HR for progression/death in the non-CMR group was significantly increased (HR, 2.94; 95% CI, 1.60-5.40; P = 0.0005), even when adjusting for LDH (HR, 2.65; 95% CI, 1.43-4.91; P = 0.0020). At week 7, 18 F-FDG PET/CT was false-positive in all 4 (6%) patients with new FDG-avid lesions but CMR of known metastases. When 18 F-FDG PET/CT was performed at progressive disease, 18/22 (82%) patients had progression of known metastases with or without new 18 F-FDG-avid lesions., Conclusions: This study shows that PERCIST response assessment at week 7 is predictive for PFS, regardless of LDH. At 2 weeks, patients with CMR have longer PFS than patients with non-CMR, but different PET parameters should be investigated to further evaluate the added value of early 18 F-FDG PET/CT. Disease progression on PET/CT is predominated by progression of known metastases, and new 18 F-FDG-avid lesions during BRAF/MEKi are not automatically a sign of recurrent disease., Competing Interests: Conflicts of interest and sources of funding: The REPOSIT study is supported by an unrestricted grant by Roche Medical B.V. The company has approved the design of the study and provided cobimetinib free of charge. The company has no role in collection, analysis, and interpretation of data or in writing the article. A.J.M.v.d.E. received study grant from Sanofi, Roche, Bristol Myers Squibb, TEVA, and Idera; received travel expenses coverage from MSD Oncology, Roche, Pfizer, and Sanofi; received speaker honoraria from Bristol Myers Squibb and Novartis; and is part of the advisory board of Bristol Myers Squibb, MSD Oncology, Amgen, Roche, Novartis, Sanofi, Pfizer, Ipsen, Merck, and Pierre Fabre. E.W.K. has consultancy/advisory relationships with Bristol Myers Squibb, Novartis, Merck, Pierre Fabre, Lilly, and Bayer, all paid to the institute; and received research grants not related to this article from Bristol Myers Squibb, Delcath, Novartis, and Pierre Fabre. G.A.P.H. has consultancy/advisory relationships with Amgen, Bristol Myers Squibb, Roche, MSD, Pfizer, Novartis, Sanofi, and Pierre Fabre, all paid to the institute; and received research grants from Bristol Myers Squibb and Seerave, all paid to the institute. M.J.B.A. has advisory board/consultancy honoraria from Amgen, Bristol Myers Squibb, Novartis, MSD-Merck, Merck-Pfizer, Pierre Fabre, Sanofi, Astellas, and Bayer, and research grants from Merck-Pfizer, all paid to the institute and not related to current work. F.Y.F.L.d.V. received research grant from Foundation STOPBraintumors.org , Bristol Myers Squibb, Novartis, Servier, CureVac, and EORTC, all paid to the institute. A.A.M.v.d.V. has consultancy roles (all paid to the institute) for Bristol Myers Squibb, MSD, Roche, Sanofi, Novartis, Pierre Fabre, Merck, Ipsen, Eisai, and Pfizer, all paid to the institute. J.B.A.G.H. has advisory roles for Bristol Myers Squibb, CureVac, GSK, Ipsen, Iovance Biotherapeutics, Imcyse, Merck Serono, Molecular Partners, MSD, Novartis, Pfizer, Roche, Sanofi, and Third Rock Ventures; is a member of SAB of Achilles Tx, BioNTech, Gadeta, Immunocore, Instil Bio, PokeAcell, Scenic, T-Knife, and Neogene Tx, all paid to the institute except Neogene Tx and Scenic; and received grant support from Amgen, Asher Bio, BioNTech, Bristol Myers Squibb, MSD, Novartis, and Sastra Cell Therapy, all paid to the institute. The other authors declare no conflicts of interest., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
36. Evaluation of the tolerability and safety of [ 225 Ac]Ac-PSMA-I&T in patients with metastatic prostate cancer: a phase I dose escalation study.
- Author
-
Ling SW, van der Veldt AAM, Konijnenberg M, Segbers M, Hooijman E, Bruchertseifer F, Morgenstern A, de Blois E, and Brabander T
- Subjects
- Male, Humans, Prostate-Specific Antigen, Prospective Studies, Retrospective Studies, Dipeptides adverse effects, Radioisotopes therapeutic use, Radiopharmaceuticals adverse effects, Heterocyclic Compounds, 1-Ring, Treatment Outcome, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
Background: Life expectancy of patients with metastatic castration-resistant prostate cancer (mCRPC) is still limited despite several systemic treatments. Within five years after diagnosis of primary prostate cancer, 10-20% of the patients have mCRPC and curation is not an option. Radionuclide therapy (RNT) targeted against prostate-specific membrane antigen (PSMA) emerged as a new treatment option and showed effective results in patients with mCRPC. Survival benefit after [
177 Lu]Lu-PSMA RNT has already been demonstrated in several clinical trials. However, [225 Ac]Ac-PSMA (225 Ac-PSMA) appears to be an even more promising radiopharmaceutical for the treatment of mCRPC. The use of alpha emitting radionuclides offers advantages over beta emitting radionuclides due to the high linear energy transfer effective for killing tumor cells and the limited range to reduce the radiation effects on the healthy tissue. However, these results are based on retrospective data and safety data of225 Ac-PSMA are still limited. Therefore, a prospective trial is needed to determine the optimal amount of activity that can be administered., Methods: The225 Ac-PSMA-Imaging & Therapy (I&T) trial is an investigator-initiated phase I, single-center, open label, repeated dose-escalation and expansion trial. Patient with PSMA-positive mCRPC after at least one line of chemotherapy and/or one line of nonsteroidal antiandrogen will be treated with225 Ac-PSMA-I&T in increasing amount of activity per cycle. Dose-escalation following an accelerated 3 + 3 design which allows to open the next dose-level cohort in the absence of dose limiting toxicity while the previous one is still ongoing. Up to 4 treatment cohorts will be explored including up to 3 dose-escalation cohorts and one expansion cohort where patients will be administered with the recommended dose. A total of up to 30 patients will be enrolled in this trial. All patients will be evaluated for safety. Additionally, dosimetry was performed for the patients in the dose-escalation cohorts after the first225 Ac-PSMA-I&T administration., Discussion: This trial will assess the safety and tolerability of225 Ac-PSMA-I&T in patients with mCRPC to recommend the optimal dose for the phase II trial., Trial Registration: ClinicalTrials.gov, (NCT05902247). Retrospectively registered 13 June 2023., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
37. Seasonal variation of anti-PD-1 outcome in melanoma-Results from a Dutch patient cohort.
- Author
-
Borgers JSW, Burgers FH, Schina A, Van Not OJ, van den Eertwegh AJM, Blank CU, Aarts MJB, van den Berkmortel FWPJ, de Groot JWB, Hospers GAP, Kapiteijn E, Piersma D, van Rijn RS, Boer AMS, van der Veldt AAM, Vreugdenhil G, Boers-Sonderen MJ, Wouters MWJM, Suijkerbuijk KPM, van Thienen JV, and Haanen JBAG
- Subjects
- Humans, Seasons, Survival Rate, Retrospective Studies, Melanoma genetics, Melanoma therapy
- Abstract
Despite the improved survival rates of patients with advanced stage melanoma since the introduction of ICIs, many patients do not have (long-term) benefit from these treatments. There is evidence that the exposome, an accumulation of host-extrinsic factors including environmental influences, could impact ICI response. Recently, a survival benefit was observed in patients with BRAF wild-type melanoma living in Denmark who initiated immunotherapy in summer as compared to winter. As the Netherlands lies in close geographical proximity to Denmark and has comparable seasonal differences, a Dutch validation cohort was established using data from our nationwide melanoma registry. In this study, we did not observe a similar seasonal difference in overall survival and are therefore unable to confirm the Danish findings. Validation of either the Dutch or Danish findings in (combined) patient cohorts from other countries would be necessary to determine whether this host-extrinsic factor influences the response to ICI-treatment., (© 2023 The Authors. Pigment Cell & Melanoma Research published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
38. Review - The impact of pharmacogenetics on the outcome of immune checkpoint inhibitors.
- Author
-
de Joode K, Heersche N, Basak EA, Bins S, van der Veldt AAM, van Schaik RHN, and Mathijssen RHJ
- Subjects
- Humans, Immune Checkpoint Inhibitors therapeutic use, Pharmacogenetics, Antineoplastic Agents, Immunological therapeutic use, Neoplasms drug therapy, Neoplasms genetics, Neoplasms chemically induced
- Abstract
The development of immune checkpoint inhibitors (ICIs) has a tremendous effect on the treatment options for multiple types of cancer. Nonetheless, there is a large interpatient variability in response, survival, and the development of immune-related adverse events (irAEs). Pharmacogenetics is the general term for germline genetic variations, which may cause the observed interindividual differences in response or toxicity to treatment. These genetic variations can either be single-nucleotide polymorphisms (SNPs) or structural variants, such as gene deletions, amplifications or rearrangements. For ICIs, pharmacogenetic variation in the human leukocyte antigen molecules has also been studied with regard to treatment outcome. This review presents a summary of the literature regarding the pharmacogenetics of ICI treatment, discusses the most important known genetic variations and offers recommendations on the application of pharmacogenetics for ICI treatment., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: K.J. declares travel expenses from Ipsen, outside the submitted work; A.A.M.V. reports advisory board (all paid to institution) of BMS, MSD, Merck, Pfizer, Ipsen, Eisai, Pierre Fabre, Roche, Novartis, Sanofi, all outside the submitted work. All other authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
39. Improved postprocessing of dynamic glucose-enhanced CEST MRI for imaging brain metastases at 3 T.
- Author
-
Wu Y, Derks SHAE, Wood TC, de Blois E, van der Veldt AAM, Smits M, and Warnert EAH
- Subjects
- Humans, Cross-Sectional Studies, Magnetic Resonance Imaging methods, Image Processing, Computer-Assisted methods, Glucose, Brain Neoplasms diagnostic imaging
- Abstract
Background: Dynamic glucose-enhanced (DGE) chemical exchange saturation transfer (CEST) has the potential to characterize glucose metabolism in brain metastases. Since the effect size of DGE CEST is small at 3 T (< 1%), measurements of signal-to-noise ratios are challenging. To improve DGE detection, we developed an acquisition pipeline and extended image analysis for DGE CEST on a hybrid 3-T positron emission tomography/magnetic resonance imaging system., Methods: This cross-sectional study was conducted after local ethical approval. Static Z-spectra (from -100 to 100 ppm) were acquired to compare the use of 1.2 versus 2 ppm to calculate static glucose-enhanced (glucoCEST) maps in 10 healthy volunteers before and after glucose infusion. Dynamic CEST images were acquired during glucose infusion. Image analysis was optimized using motion correction, dynamic B
0 correction, and principal component analysis (PCA) to improve the detection of DGE CEST in the sagittal sinus, cerebrospinal fluid, and grey and white matter. The developed DGE CEST pipeline was applied to four patients diagnosed with brain metastases., Results: GlucoCEST was strongest in healthy tissues at 2 ppm. Correcting for motion, B0, and use of PCA locally improved DGE maps. A larger contrast between healthy tissues and enhancing regions in brain metastases was found when dynamic B0 correction and PCA denoising were applied., Conclusion: We demonstrated the feasibility of DGE CEST with our developed acquisition and analysis pipeline at 3 T in patients with brain metastases. This work enables a direct comparison of DGE CEST to 18F-fluoro-deoxy-D-glucose positron emission tomography of glucose metabolism in patients with brain metastases., Relevance Statement: Contrast between brain metastasis and healthy brain tissue in DGE CEST MR images is improved by including principle component analysis and dynamic magnetic field correction during postprocessing. This approach enables the detection of increased DGE CEST signal in brain metastasis, if present., Key Points: • Despite the low signal-to-noise ratio, dynamic glucose-enhanced CEST MRI is feasible at 3 T. • Principal component analyses and dynamic magnetic field correction improve DGE CEST MRI. • DGE CEST MRI does not consequently show changes in brain metastases compared to healthy brain tissue. • Increased DGE CEST MRI in brain metastases, if present, shows overlap with contrast enhancement on T1-weighted images., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
40. Clinical evaluation of the clinicopathologic and gene expression profile (CP-GEP) in patients with melanoma eligible for sentinel lymph node biopsy: A multicenter prospective Dutch study.
- Author
-
Stassen RC, Mulder EEAP, Mooyaart AL, Francken AB, van der Hage J, Aarts MJB, van der Veldt AAM, Verhoef C, and Grünhagen DJ
- Subjects
- Humans, Sentinel Lymph Node Biopsy, Transcriptome, Prospective Studies, Lymphatic Metastasis pathology, Neoplasm Staging, Melanoma genetics, Melanoma surgery, Melanoma pathology, Skin Neoplasms genetics, Skin Neoplasms surgery, Skin Neoplasms diagnosis, Sentinel Lymph Node pathology
- Abstract
Sentinel lymph node biopsy (SLNB) is recommended for patients with >pT1b cutaneous melanoma, and should be considered and discussed with patients diagnosed with pT1b cutaneous melanoma for the purpose of staging, prognostication and determining eligibility for adjuvant therapy. Previously, the clinicopathologic and gene expression profile (CP-GEP, Merlin Assay®) model was developed to identify patients who can forgo SLNB because of a low risk for sentinel node metastasis. The aim of this study was to evaluate the clinical use and implementation of the CP-GEP model in a prospective multicenter study in the Netherlands. Both test performance and feasibility for clinical implementation were assessed in 260 patients with T1-T4 melanoma. The CP-GEP model demonstrated an overall negative predictive value of 96.7% and positive predictive value of 23.7%, with a potential SLNB reduction rate of 42.2% in patients with T1-T3 melanoma. With a median time of 16 days from initiation to return of test results, there was sufficient time left before the SLNB was performed. Based on these outcomes, the model may support clinical decision-making to identify patients who can forgo SLNB in clinical practice., Competing Interests: Declaration of competing interest This study was partially funded by SkylineDx. All other authors declare that they have no potential or competing interests., (© 2023 Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
41. Association between pretreatment emotional distress and neoadjuvant immune checkpoint blockade response in melanoma.
- Author
-
Fraterman I, Reijers ILM, Dimitriadis P, Broeks A, Gonzalez M, Menzies AMM, Lopez-Yurda M, Kapiteijn E, van der Veldt AAM, Suijkerbuijk KPM, Hospers GAP, Long GV, Blank CU, and van de Poll-Franse LV
- Subjects
- Humans, Immune Checkpoint Inhibitors therapeutic use, Neoadjuvant Therapy, Glucocorticoids therapeutic use, Neoplasm Recurrence, Local pathology, Biomarkers, Tumor genetics, Interferon-gamma therapeutic use, Adrenergic Agents therapeutic use, Melanoma drug therapy, Melanoma genetics, Melanoma pathology, Psychological Distress
- Abstract
Neoadjuvant immune checkpoint blockade (ICB) outperforms adjuvant ICB for treatment of stage IIIB-D melanoma, but potential biomarkers of response, such as interferon-gamma (IFNγ) signature and tumor mutational burden (TMB), are insufficient. Preclinical studies suggest that emotional distress (ED) can negatively affect antitumor immune responses via β-adrenergic or glucocorticoid signaling. We performed a post hoc analysis evaluating the association between pretreatment ED and clinical responses after neoadjuvant ICB treatment in patients with stage IIIB-D melanoma in the phase 2 PRADO trial ( NCT02977052 ). The European Organisation for Research and Treatment of Cancer scale for emotional functioning was used to identify patients with ED (n = 28) versus those without (n = 60). Pretreatment ED was significantly associated with reduced major pathologic responses (46% versus 65%, adjusted odds ratio 0.20, P = 0.038) after adjusting for IFNγ signature and TMB, reduced 2-year relapse-free survival (74% versus 91%, adjusted hazard ratio 3.81, P = 0.034) and reduced 2-year distant metastasis-free survival (78% versus 95%, adjusted hazard ratio 4.33, P = 0.040) after adjusting for IFNγ signature. RNA sequencing analyses of baseline patient samples could not identify clear β-adrenergic- or glucocorticoid-driven mechanisms associated with these reduced outcomes. Pretreatment ED may be a marker associated with clinical responses after neoadjuvant ICB in melanoma and warrants further investigation. ClinicalTrials.gov registration: NCT02977052 ., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
42. 68 Ga-PSMA PET/CT for Response Evaluation of 223 Ra Treatment in Metastatic Prostate Cancer.
- Author
-
de Jong AC, Segbers M, Ling SW, Graven LH, Mehra N, Hamberg P, Brabander T, de Wit R, and van der Veldt AAM
- Subjects
- Male, Humans, Positron Emission Tomography Computed Tomography methods, Prospective Studies, Treatment Outcome, Gallium Radioisotopes therapeutic use, Retrospective Studies, Prostate-Specific Antigen, Prostatic Neoplasms, Castration-Resistant diagnostic imaging, Prostatic Neoplasms, Castration-Resistant radiotherapy, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms therapy, Prostatic Neoplasms pathology
- Abstract
CT and bone scintigraphy are not useful for response evaluation of bone metastases to
223 Ra treatment in metastatic castration-resistant prostate cancer (mCRPC). PET using68 Ga prostate-specific membrane antigen 11 (68 Ga-PSMA) is a promising tool for response evaluation of mCRPC. The aim of this study was to determine the utility of68 Ga-PSMA PET/CT for response evaluation of223 Ra treatment in patients with mCRPC. Methods: Within this prospective, multicenter, imaging discovery study, 28 patients with mCRPC, eligible for223 Ra treatment, were included between 2019 and 2022. Patients received223 Ra according to the standard of care. Study procedures included CT, bone scintigraphy, and68 Ga-PSMA PET/CT at baseline, after 3 and 6 cycles of223 Ra treatment, and on treatment failure. Response to223 Ra treatment was visually assessed on all 3 imaging modalities. Total tumor volume within bone (TTVbone ) was determined on68 Ga-PSMA PET/CT. Intrapatient heterogeneity in response was studied using a newly developed image-registration tool for sequential images of PET/CT. Results were compared with failure-free survival (good responders vs. poor responders; cutoff, 24 wk) and alkaline phosphatase (ALP) response after 3 cycles. Results: Visual response assessment criteria could not distinguish good responders from poor responders on68 Ga-PSMA PET/CT and bone scintigraphy. For68 Ga-PSMA PET/CT, TTVbone at baseline was lower in good responders than in poor responders, whereas TTVbone increased in both groups during treatment. TTVbone was higher in patients with new extraosseous metastases during223 Ra treatment. Although TTVbone and ALP correlated at baseline, changes in TTVbone and ALP on treatment did not.68 Ga-PSMA response of TTVbone showed intrapatient heterogeneity in most patients. Conclusion: mCRPC patients with lower TTVbone on68 Ga-PSMA PET/CT have the best clinical outcome after223 Ra treatment. Response is highly heterogeneous in most patients. A decrease in ALP, which occurred in most patients, was not correlated with a decrease in TTVbone , which might make one question the value of ALP for disease monitoring during223 Ra treatment in clinical practice., (© 2023 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2023
- Full Text
- View/download PDF
43. Liquid Biopsies for Early Response Evaluation of Radium-223 in Metastatic Prostate Cancer.
- Author
-
de Jong AC, Isebia KT, Ling SW, de Weerd V, Van NM, Kraan J, Martens JWM, Mehra N, Hamberg P, Lolkema MP, de Wit R, van der Veldt AAM, and Wilting SM
- Subjects
- Male, Humans, Prospective Studies, Biomarkers, Tumor genetics, Liquid Biopsy, Aneuploidy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant radiotherapy, Prostatic Neoplasms, Castration-Resistant drug therapy, Cell-Free Nucleic Acids
- Abstract
Purpose: Reliable biomarkers for response monitoring during radium-223 treatment in patients with metastatic castration-resistant prostate cancer (mCRPC) are lacking. Circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA), obtained from liquid biopsies, are shown to have prognostic value in mCRPC. The aim of this study was to determine the value of CTCs and ctDNA for response evaluation of radium-223., Methods: In this prospective multicenter study, longitudinal blood draws and imaging were performed in 28 patients with mCRPC and predominantly bone disease, who were treated with radium-223. CTCs were counted (CELLSEARCH CTC test), while fraction of ctDNA was estimated by measuring aneuploidy of cell-free DNA (cfDNA; modified Fast Aneuploidy Screening Test-Sequencing System). CTC counts and aneuploidy score (AS) were categorized as low (<5) and high (≥5). Primary and secondary clinical end points were failure-free survival (FFS), and overall survival (OS) and development of extraosseous metastases, respectively. Additionally, CTC count and AS were related to alkaline phosphatase (ALP) and total tumor volume in bone (TTV
bone ) on positron emission tomography-computed tomography with68 gallium prostate-specific membrane antigen., Results: FFS was longer in patients with a low CTC count or AS either at baseline or after 12 weeks, whereas for OS, only a significant association with CTC count was observed. Liquid biopsy results correlated well with ALP and TTVbone at baseline, but not with change in both parameters after three cycles of radium-223. AS and CTC count were significantly correlated., Conclusion: CTC count and AS of cfDNA at baseline and during treatment predict clinical response to radium-223 in patients with mCRPC, warranting future evaluation of their value in treatment guidance.- Published
- 2023
- Full Text
- View/download PDF
44. Factors associated with long-term antibody response after COVID-19 vaccination in patients treated with systemic treatment for solid tumors.
- Author
-
Oosting SF, van der Veldt AAM, Fehrmann RSN, Bhattacharya A, van Binnendijk RS, GeurtsvanKessel CH, Dingemans AC, Smit EF, Hiltermann TJN, den Hartog G, Jalving M, Westphal TT, de Wilt F, Ernst SM, Boerma A, van Zijl L, Rimmelzwaan GF, Kvistborg P, van Els CACM, Rots NY, van Baarle D, Haanen JBAG, and de Vries EGE
- Subjects
- Humans, COVID-19 Vaccines, Antibody Formation, Vaccination, COVID-19, Neoplasms drug therapy
- Published
- 2023
- Full Text
- View/download PDF
45. Adjuvant treatment of in-transit melanoma: Narrowing the knowledge gap left by clinical trials.
- Author
-
de Meza MM, Blokx WAM, Bonenkamp HJ, Blank CU, Aarts MJB, van den Berkmortel FWPJ, Boers-Sonderen MJ, de Groot JWB, Haanen JB, Hospers GAP, Kapiteijn EW, van Not OJ, Piersma D, van Rijn RS, Stevense-Den Boer MA, van der Veldt AAM, Vreugdenhil G, van den Eertwegh AJM, Suijkerbuijk KPM, and Wouters MWJM
- Subjects
- Humans, Combined Modality Therapy, Melanoma, Cutaneous Malignant, Melanoma pathology, Skin Neoplasms pathology
- Abstract
Few clinical trials address efficacy of adjuvant systemic treatment in patients with in-transit melanoma (ITM). This study describes adjuvant systemic therapy of ITM patients beyond clinical trials. In this study, we included stage III adjuvant-treated melanoma patients registered in the nationwide Dutch Melanoma Treatment Registry between July 2018 and December 2020. Patients were divided into three groups: nodal disease only, ITM only and ITM and nodal disease. Recurrence patterns, recurrence-free survival (RFS) and overall survival (OS) at 12-months were analyzed. In our study population of 1037 patients, 66.8% had nodal disease only, 16.7% had ITM only and 16.2% had ITM with nodal disease. RFS at 12-months was comparable in the nodal only and ITM only group (72.2% vs70.1%, P = .97) but lower in ITM and nodal disease patients (57.8%; P = .01, P < .01). Locoregional metastases occurred as first recurrence in 38.9% nodal disease only, 71.9% of ITM-only and 44.0% of ITM and nodal disease patients. Distant recurrences occurred in 42.3%, 18.8% and 36.0%, respectively (P = .02). 12-months OS was not significantly different for nodal disease only patients compared with ITM-only (94.4% vs 97.6%, P = .06) but was significantly higher for ITM-only compared with ITM and nodal disease patients (97.6% vs 91.0%, P < .01). In conclusion, we showed that in the adjuvant setting, RFS rates in ITM-only patients are similar to non-ITM, though better than in ITM and nodal disease patients. Adjuvant-treated ITM-only patients less often experience distant recurrences and have a superior OS compared with ITM and nodal disease patients., (© 2023 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2023
- Full Text
- View/download PDF
46. The genomic and transcriptomic landscape of advanced renal cell cancer for individualized treatment strategies.
- Author
-
de Joode K, van de Geer WS, van Leenders GJLH, Hamberg P, Westgeest HM, Beeker A, Oosting SF, van Rooijen JM, Beerepoot LV, Labots M, Mathijssen RHJ, Lolkema MP, Cuppen E, Sleijfer S, van de Werken HJG, and van der Veldt AAM
- Subjects
- Humans, Transcriptome, Prospective Studies, Genomics, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology
- Abstract
Differences in the clinical course and treatment responses in individual patients with advanced renal cell carcinoma (RCC) can largely be explained by the different genomics of this disease. To improve the personalized treatment strategy and survival outcomes for patients with advanced RCC, the genomic make-up in patients with advanced RCC was investigated to identify putative actionable variants and signatures. In this prospective multicenter study (NCT01855477), whole-genome sequencing (WGS) data of locally advanced and metastatic tissue biopsies and matched whole-blood samples were collected from 91 patients with histopathologically confirmed RCC. WGS data were analyzed for small somatic variants, copy-number alterations and structural variants. For a subgroup of patients, RNA sequencing (RNA-Seq) data could be analyzed. RNA-Seq data were clustered on immunogenic and angiogenic gene expression patterns according to a previously developed angio-immunogenic gene signature. In all patients with papillary and clear cell RCC, putative actionable drug targets were detected by WGS, of which 94% were on-label available. RNA-Seq data of clear cell and papillary RCC were clustered using a previously developed angio-immunogenic gene signature. Analyses of driver mutations and RNA-Seq data revealed clear differences among different RCC subtypes, showing the added value of WGS and RNA-Seq over clinicopathological data. By improving both histological subtyping and the selection of treatment according to actionable targets and immune signatures, WGS and RNA-Seq may improve therapeutic decision making for most patients with advanced RCC, including patients with non-clear cell RCC for whom no standard treatment is available to data. Prospective clinical trials are needed to evaluate the impact of genomic and transcriptomic diagnostics on survival outcome for advanced RCC patients., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
47. Time interval from primary melanoma to first distant recurrence in relation to patient outcomes in advanced melanoma.
- Author
-
van Duin IAJ, Elias SG, van den Eertwegh AJM, de Groot JWB, Blokx WAM, van Diest PJ, Leiner T, Verhoeff JJC, Verheijden RJ, van Not OJ, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Haanen JBAG, Hospers GAP, Kamphuis AM, Piersma D, van Rijn RS, van der Veldt AAM, Vreugdenhil G, Wouters MWJM, Stevense-den Boer MAM, Boers-Sonderen MJ, Kapiteijn E, and Suijkerbuijk KPM
- Subjects
- Humans, Prognosis, Antineoplastic Combined Chemotherapy Protocols adverse effects, Mitogen-Activated Protein Kinase Kinases, Retrospective Studies, Proto-Oncogene Proteins B-raf genetics, Melanoma
- Abstract
Since the introduction of BRAF(/MEK) inhibition and immune checkpoint inhibition (ICI), the prognosis of advanced melanoma has greatly improved. Melanoma is known for its remarkably long time to first distant recurrence (TFDR), which can be decades in some patients and is partly attributed to immune-surveillance. We investigated the relationship between TFDR and patient outcomes after systemic treatment for advanced melanoma. We selected patients undergoing first-line systemic therapy for advanced melanoma from the nationwide Dutch Melanoma Treatment Registry. The association between TFDR and progression-free survival (PFS) and overall survival (OS) was assessed by Cox proportional hazard regression models. The TFDR was modeled categorically, linearly, and flexibly using restricted cubic splines. Patients received anti-PD-1-based treatment (n = 1844) or BRAF(/MEK) inhibition (n = 1618). For ICI-treated patients with a TFDR <2 years, median OS was 25.0 months, compared to 37.3 months for a TFDR >5 years (P = .014). Patients treated with BRAF(/MEK) inhibition with a longer TFDR also had a significantly longer median OS (8.6 months for TFDR <2 years compared to 11.1 months for >5 years, P = .004). The hazard of dying rapidly decreased with increasing TFDR until approximately 5 years (HR 0.87), after which the hazard of dying further decreased with increasing TFDR, but less strongly (HR 0.82 for a TFDR of 10 years and HR 0.79 for a TFDR of 15 years). Results were similar when stratifying for type of treatment. Advanced melanoma patients with longer TFDR have a prolonged PFS and OS, irrespective of being treated with first-line ICI or targeted therapy., (© 2023 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2023
- Full Text
- View/download PDF
48. Real-world Outcomes of Ipilimumab Plus Nivolumab Combination Therapy in a Nation-wide Cohort of Advanced Melanoma Patients in the Netherlands.
- Author
-
van Zeijl MCT, van Breeschoten J, de Wreede LC, Wouters MWJM, Hilarius DL, Blank CU, Aarts MJB, van den Berkmortel FWPJ, de Groot JWB, Hospers GAP, Kapiteijn E, Piersma D, van Rijn RS, Stevense-den Boer MA, van der Veldt AAM, Vreugdenhil G, Boers-Sonderen MJ, Suijkerbuijk KPM, Haanen JBAG, and van den Eertwegh AJM
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Ipilimumab therapeutic use, Netherlands, Nivolumab therapeutic use, Brain Neoplasms drug therapy, Melanoma pathology
- Abstract
In phase III trials, ipilimumab plus nivolumab combination therapy is highly efficacious for advanced melanoma, despite many treatment-related grades 3-4 adverse events. Here, we report real-world safety and survival outcomes of ipilimumab plus nivolumab for advanced melanoma. Patients with advanced melanoma who received first-line ipilimumab plus nivolumab between January 1, 2015 and June 30, 2021 were selected from the Dutch Melanoma Treatment Registry. We evaluated response status at 3, 6, 12, 18, and 24 months. OS and PFS were estimated with the Kaplan-Meier method. Separate analyses were performed for patients with or without brain metastases and for patients who met the inclusion criteria of the Checkmate-067 trial. In total, 709 patients received first-line ipilimumab plus nivolumab. Three hundred sixty (50.7%) patients experienced grade 3-4 adverse events, with 211 of the (58.6%) patients requiring hospital admission. The median treatment duration was 42 days (IQR = 31-139). At 24 months, disease control was achieved in 37% of patients. Median PFS since the start of treatment was 6.6 months (95% CI: 5.3-8.7), and median OS was 28.7 months (95% CI: 20.7-42.2). CheckMate-067 trial-like patients had a 4-year OS of 50% (95% CI: 43-59). Among patients with no asymptomatic or symptomatic brain metastases, the 4-year OS probabilities were 48% (95% CI: 41-55), 45% (95% CI: 35-57), and 32% (95% CI: 23-46). Ipilimumab plus nivolumab can achieve long-term survival in advanced melanoma patients in a real-world setting, including patients not represented in the CheckMate-067 trial. However, the proportion of patients with disease control in the real world is lower compared with clinical trials., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
49. Response to checkpoint inhibition and targeted therapy in melanoma patients with concurrent haematological malignancies.
- Author
-
Van Not OJ, van den Eertwegh AJM, Haanen JB, van Rijn RS, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, van Eijs MJM, de Groot JB, Hospers GAP, Kapiteijn E, de Meza M, Piersma D, Stevense-den Boer M, van der Veldt AAM, Vreugdenhil G, Wouters MWJM, Suijkerbuijk KPM, and Blokx WAM
- Subjects
- Humans, Nivolumab therapeutic use, Ipilimumab, Prospective Studies, Proto-Oncogene Proteins B-raf, Retrospective Studies, Mitogen-Activated Protein Kinase Kinases, Melanoma pathology, Hematologic Neoplasms
- Abstract
Background: Patients diagnosed with haematologic malignancies (HMs) have a higher risk of developing subsequent solid tumours, such as melanoma. Patients with HM were mostly excluded from clinical trials but potentially derive less benefit from immune checkpoint inhibitors (ICIs) due to disease- or treatment-related T- or B-cell dysfunction., Methods: All advanced melanoma patients treated with anti-PD-1-based treatment or targeted therapy between 2015 and 2021 were included from the prospective nationwide Dutch Melanoma Treatment Registry. Progression-free survival (PFS) and melanoma-specific survival (MSS) were analysed for patients with HM (HM+) and without HM (HM-). A cox model was used to account for confounders associated with PFS and MSS., Results: In total, 4638 advanced melanoma patients received first-line anti-PD-1 monotherapy (n = 1763), ipilimumab-nivolumab (n = 800), or BRAF(/MEK) inhibitors (n = 2075). Concurrent HMs were present for 46 anti-PD1-treated patients, 11 ipilimumab-nivolumab-treated patients and 43 BRAF(/MEK)-inhibitor-treated patients. In anti-PD-1-treated patients, the median PFS was 2.8 months for HM+ and 9.9 months for HM- (p = 0.01). MSS was 41.2 months for HM+ and 58.1 months for HM- (p = 0.00086). In multivariable analysis, the presence of an HM was significantly associated with higher risk of melanoma progression (HR
adj 1.62; 95% confidence interval [95% CI] 1.15-2.29; p = 0.006) and melanoma-related death (HRadj 1.74; 95% CI 1.09-2.78; p = 0.020). Median PFS and MSS for first-line BRAF(/MEK-) inhibitor-treated HM+ and HM- patients were not significantly different., Conclusions: Patients with HM and advanced melanoma show significantly worse melanoma-related outcomes when treated with ICI, but not targeted therapy, compared to patients without HM. Clinicians should be aware of potentially altered effectiveness of ICI in patients with active HM., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: A.vdE. has advisory relationships with Amgen, Bristol Myers Squibb, Roche, Novartis, MSD, Pierre Fabre, Sanofi, Pfizer, Ipsen, Merck and has received research study grants not related to this paper from Sanofi, Roche, Bristol Myers Squibb, Idera and TEVA and has received travel expenses from MSD Oncology, Roche, Pfizer and Sanofi and has received speaker honoraria from BMS and Novartis. J.H. has advisory relationships with Achilles Therapeutics, AstraZeneca, Bristol Myers Squibb, BioNTech, Immunocore, Iovance Biotherapeutics, Instil Bio, Ipsen, MSD, Merck Serono, Molecular Partners, Novartis, Neogene Therapeutics, Pfizer, PokeAcel, Roche/Genentech, Sanofi, T-Knife and has received research grants not related to this paper from Asher Bio, Amgen, Bristol Myers Squibb, MSD, BioNTech, Neogene Therapeutics and Novartis. All grants were paid to the institutions. C.B. has/had advisory role: BMS, MSD, Roche, Novartis, GSK, AZ, Pfizer, Lilly, GenMab, Pierre Fabre, Third Rock Ventures, received research funding: BMS, Novartis, NanoString, 4SC, stockownership: co-founder Immagene BV and Signature Oncology, patents (incl. submitted): WO 2021/177822 A1, N2027907, P091040NL2. M.A. has advisory board/consultancy honoraria from Amgen, Bristol Myers Squibb, Novartis, MSD-Merck, Merck-Pfizer, Pierre Fabre, Sanofi, Astellas, Bayer. Research grants Merck-Pfizer. Not related to current work and paid to institute. J.dG. has consultancy/advisory relationships with Bristol Myers Squibb, Pierre Fabre, Servier, MSD, Novartis. G.H. consultancy/advisory relationships with Amgen, Bristol Myers Squibb, Roche, MSD, Pfizer, Novartis, Pierre Fabre and has received research grants not related to this paper from Bristol Myers Squibb, Seerave and were paid to the institution. E.K. has consultancy/advisory relationships with Bristol Myers Squibb, Novartis, Merck, Pierre Fabre, Lilly and Bayer, and received research grants not related to this paper from Bristol Myers Squibb and Pierre-Fabre. Not related to current work and paid to institute. R.vR. has advisory board/consultancy honoraria from Pfizer and an expert meeting fee from Roche. A.vdV. has consultancy relationships with Bristol Myers Squibb, MSD, Roche, Novartis, Pierre Fabre, Pfizer, Sanofi, Ipsen, Eisai, Merck. M.B.S. has consultancy/advisory relationships with Pierre Fabre, MSD and Novartis. K.S. has advisory relationships with Bristol Myers Squibb, Novartis, MSD, Pierre Fabre, AbbVie, received honoraria from Novartis, MSD and Roche and received research funding from BMS, Philips and TigaTx. All remaining authors have declared no conflicts of interest., (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
50. A Survival Tree of Advanced Melanoma Patients with Brain Metastases Treated with Immune Checkpoint Inhibitors.
- Author
-
van Not OJ, Wind TT, Ismail RK, Bhattacharya A, Jalving M, Blank CU, Aarts MJB, van den Berkmortel FWPJ, Boers-Sonderen MJ, van den Eertwegh AJM, de Groot JWB, Haanen JB, Kapiteijn E, Bloem M, Piersma D, van Rijn RS, Stevense-den Boer M, van der Veldt AAM, Vreugdenhil G, Wouters MWJM, Blokx WAM, Suijkerbuijk KPM, Fehrmann RSN, and Hospers GAP
- Abstract
The efficacy of immune checkpoint inhibitors (ICIs) in patients with advanced melanoma that develop brain metastases (BM) remains unpredictable. In this study, we aimed to identify prognostic factors in patients with melanoma BM who are treated with ICIs. Data from advanced melanoma patients with BM treated with ICIs in any line between 2013 and 2020 were obtained from the Dutch Melanoma Treatment Registry. Patients were included from the time of the treatment of BM with ICIs. Survival tree analysis was performed with clinicopathological parameters as potential classifiers and overall survival (OS) as the response variable. In total, 1278 patients were included. Most patients were treated with ipilimumab-nivolumab combination therapy (45%). The survival tree analysis resulted in 31 subgroups. The median OS ranged from 2.7 months to 35.7 months. The strongest clinical parameter associated with survival in advanced melanoma patients with BM was the serum lactate dehydrogenase (LDH) level. Patients with elevated LDH levels and symptomatic BM had the worst prognosis. The clinicopathological classifiers identified in this study can contribute to optimizing clinical studies and can aid doctors in giving an indication of the patients' survival based on their baseline and disease characteristics.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.