Skip to main content

Open Access 06.05.2024 | REVIEW

Past, Present, and Future Therapeutic Strategies for NF-1-Associated Tumors

verfasst von: Brian Na, Shilp R. Shah, Harish N. Vasudevan

Erschienen in: Current Oncology Reports

Abstract

Purpose of Review

Neurofibromatosis type 1 (NF-1) is a cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene that encodes the neurofibromin protein, which functions as a negative regulator of Ras signaling. We review the past, current, and future state of therapeutic strategies for tumors associated with NF-1.

Recent Findings

Therapeutic efforts for NF-1-associated tumors have centered around inhibiting Ras output, leading to the clinical success of downstream MEK inhibition for plexiform neurofibromas and low-grade gliomas. However, MEK inhibition and similar molecular monotherapy approaches that block Ras signaling do not work for all patients and show limited efficacy for more aggressive cancers such as malignant peripheral nerve sheath tumors and high-grade gliomas, motivating novel treatment approaches.

Summary

We highlight the current therapeutic landscape for NF-1-associated tumors, broadly categorizing treatment into past strategies for serial Ras pathway blockade, current approaches targeting parallel oncogenic and tumor suppressor pathways, and future avenues of investigation leveraging biologic and technical innovations in immunotherapy, pharmacology, and gene delivery.
Hinweise

Publisher's Note

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

Introduction

Neurofibromatosis type 1 (NF-1) is an autosomal dominant genetic disorder affecting 1 in 3000 individuals caused by germline mutation of the NF1 gene. The NF1 gene product neurofibromin is a Ras GTPase-activating protein (RAS-GAP) that converts active GTP-bound Ras into inactive GDP-bound Ras [14]. Thus, NF1 loss leads to constitutive Ras activation and many clinical manifestations of NF-1 such as café-au-lait macules, seizures, chronic pain, vascular issues, bone defects, central and peripheral nervous system tumors, breast cancer, and other malignancies [5, 6]. Of note, tumorigenesis typically requires a second somatic hit and consequent loss of function in the remaining wildtype NF1 allele [7].
Patients with NF-1 are at significantly increased risk for plexiform neurofibromas (PNs), a benign peripheral nervous system tumor that can transform into malignant peripheral nerve sheath tumors (MPNSTs), and low-grade gliomas (LGGs), a benign central nervous system tumor that can transform into malignant high-grade gliomas (HGGs) [810]. In addition, atypical neurofibromatous neoplasms of uncertain biologic potential (ANNUBP) comprise an intermediate tumor entity that reflect the transition from plexiform neurofibromas to MPNSTs [11]. ANNUBPs are associated with CDKN2AB loss, and their diagnosis and classification remain an area of active investigation [12•, 13, 14].
Here, we summarize past, present, and future treatment approaches for NF1-associated tumors. Given neurofibromin’s function as a Ras-GAP and resulting Ras pathway misactivation, therapies to date have primarily focused on inhibiting Ras signaling output at the level of RAF, MEK, ERK, and mTOR [15]. Leveraging our improved understanding of additional genetic hits required for NF1-associated tumorigenesis, more recent work leverages novel pharmacologic approaches to block parallel pathways such as PRC2 or CDKN2A/B loss. We conclude with an eye toward the future of NF1 therapeutics currently in preclinical development and early clinical trials including oncolytic viruses, cellular therapy, immune checkpoint inhibitors, gene therapy, and direct Ras inhibition.

Past Approaches: Serial Inhibition Along the Ras Signaling Axis

Ras signaling begins at the cell membrane with receptor tyrosine kinase (RTK) activation, setting off a signaling cascade to activate Ras through G-protein exchange factors (GEFs) such as SOS, a process that requires SHP2 and adapter proteins such as GRB2 to promote the formation of active GTP bound Ras (Fig. 1A) [16]. GTP-bound Ras subsequently activates RAF-MEK-ERK while mTOR is classically activated by PI3K signaling, classically through RTK activation with a potential contribution directly by active GTP-bound Ras. Accordingly, upstream RTKs and downstream RAF-MEK-ERK and mTOR have been the primary area of therapeutic investigation to date.

Mitogen-Activated Protein Kinase Kinase (MEK) Inhibitors (MEKi)

MEK inhibitors (MEKi) have shown significant efficacy for NF1-associated PNs. In particular, the MEKi selumetinib has received FDA approval for symptomatic and inoperable PNs in patients aged 2–18, with ongoing Phase 2 trials in adults displaying similar positive responses [17, 18•, 19]. The success has spurred the investigation of other MEKi such as binimetinib, cobimetinib, and mirdametinib to enhance clinical efficacy and impact on the tumor microenvironment [20].
Beyond PNs, MEKi is under investigation for additional NF1 manifestations, including atypical neurofibromas, MPNSTs, cutaneous neurofibromas, LGGs, and juvenile myelomonocytic leukemia. In particular, MEKi for cutaneous neurofibromas and LGGs is currently being tested in Phase 2 and 3 trials (NCT03871257, NCT03363217, NCT02285439, NCT03326388, NCT04201457) [21]. Beyond tumor-associated manifestations, MEKi may also have utility for non-tumor manifestations such as pain, bone issues, and neurocognition [17, 18•, 22, 23]. Clinical trials indicate reduced pain in PNs, hinting at the MEK pathway’s role in NF1-related pain and suggesting that the tumor microenvironment plays an instrumental function in NF1-PN pathogenesis [17, 18•, 2325]. Despite MEKi’s promise, many challenges remain including dosing strategies, heterogenous responses, treatment resistance, and long-term safety persist, underscoring the need for additional research.

RAF Inhibition

RAF inhibition, the most proximal downstream signaling protein from RAS, has been studied extensively in NF1-mutant tumors. First-generation RAF inhibitors selectively targeting the BRAFV600E mutation show minimal efficacy, with resistance occurring within 6 to 7 months [26, 27]. Accordingly, pan-RAF inhibitors have been developed to address these challenges. Tovorafenib, which inhibits both monomeric and dimeric BRAF, has demonstrated efficacy in pre-clinical NF1 mutant glioma models, and building on these promising results, a Phase 2 trial FIREFLY-1 (NCT04775485) investigated tovorafenib for recurrent pediatric LGGs and demonstrated a meaningful radiographic response, albeit not exclusively in NF1-mutant LGG [28•].

ERK Inhibition

The ERK inhibitor ulixertinib, a novel first-in-class drug exhibiting highly selective, reversible ATP-competitive inhibition of ERK1/2, has demonstrated an antitumor profile for MAPK-activated LGGs [29], and multiple clinical trials testing ulixertinib in the context of NF1-deficient cancers are currently underway (NCT05804227, NCT03454035). In addition, preclinical work in mice suggests ERK inhibition may be effective as combination therapy for plexiform neurofibromas [30]. MK-8353 is another ERK1/2 inhibitor that targets both the active and inactive form of ERK [31], but an open-label phase 1b clinical trial investigating the combination therapy of MK-8353 with MEKi selumetinib for advanced solid tumors found unacceptable levels of toxicity at dose levels required for clinical response (NCT03745989) [32]. Additionally, concern has been raised over the long-term effects of both MEK and ERK inhibition on abnormal skeletal manifestations inherent to NF-1 patients. In that regard, the tyrosine kinase inhibitor ponatinib with activity against MEKK2 rescues skeletal defects in vivo, perhaps offering an additional combinatorial strategy to optimize the therapeutic window of ERK inhibitors [33].

Tyrosine Kinase Inhibitors (TKIs) and SHP2 Inhibition

TKIs disrupt upstream RTK input into Ras signaling, and initial studies with the multi-TKI sunitinib showed reduced tumor burden in a mouse model of NF1-related PNs [34, 35]. However, a subsequent clinical trial was terminated following an adverse event [36]. Furthermore, trials for TKIs imatinib and sorafenib exhibited only modest efficacy in PNs [37, 38]. A more recent Phase 2 trial with the TKI cabozantinib showed promise, with 42% of participants achieving a partial response in progressive PNs [39]. It remains unclear if this effect is mediated directly through RTKs or via alternate pathways, as preclinical work indicates that cabozantinib activity against MAPK interacting kinases (MNKs), when combined with the MEKi mirdametinib, induces regression in a genetically engineered mouse malignant peripheral nerve sheath tumor (MPNST) model [40].
Another approach to modulate upstream inputs is through SHP2, which potentiates Ras GTP loading, and thus, SHP2 inhibitors (SHP2i) may offer a promising approach for NF-1-associated tumors [41]. Indeed, NF1-mutant neuroblastomas are sensitive to the SHP2 inhibitor SHP099, and the combination of MEKi/SHP2i demonstrated improved efficacy across multiple preclinical models [42, 43]. By targeting signaling proteins upstream within the RAS-MAPK pathway, SHP2 inhibition may potentiate other targeted therapies in NF-1-associated tumors.

Mammalian Target of Rapamycin (mTOR) Inhibitors

The mTOR pathway is hyperactivated in NF1-deficient tumors [8, 44, 45]. Sirolimus, an FDA-approved mTOR inhibitor, was tested for PNs in a Phase 2 clinical trial, leading to increased time to progression but no significant difference in tumor volume [46, 47]. Similarly, everolimus was studied in a Phase 2 trial and showed no efficacy for NF1-related PNs but exhibited a significant radiographic reduction in recurrent NF1-LGGs, perhaps underscoring heterogeneity between different NF1 tumor entities [48, 49]. A recently completed Phase 2 trial investigated a combination therapy of sirolimus plus MEKi for unresectable or metastatic MPNSTs, and final trial results are eagerly anticipated [50].

Present Approaches: Parallel Inhibition of Co-mutated Tumor Suppressors

Following NF1 loss, additional genetic hits are required for malignant transformation of benign nervous system tumors into their malignant entities [51, 52]. Of these, CDKN2A/B loss and PRC2 loss are well-appreciated steps in the transition from PN to MPNST [53, 54]. Loss of the tumor suppressor CDKN2A/B, which is associated with transition from PN to ANNUBP, leads to cyclin-dependent kinase (CDK) activation, motivating the application of CDK4/6 inhibitors (CDK4/6i) in NF1-associated tumors [55]. With respect to PRC2, SUZ12 and EED, obligate members of the PRC2 epigenetic complex, are recurrently mutated in MPNSTs but not PNs [54].

CDK4/6 Inhibition (CDK4/6i)

The CDK4/6i abemaciclib demonstrated synergistic anti-tumor effects when combined with the ERKi LY3214996 for PN treatment in vivo [30]. A clinical trial (NCT04000529) is ongoing to evaluate the safety and efficacy of ribociclib combined with the SHP2 inhibitor TNO155 for advanced solid tumors. In NF1-mutant breast cancer, the CDK4/6i palbociclib reduced growth and enhanced sensitivity to the antiestrogenic medication fulvestrant, indicating a synergistic relationship [56, 57]. These findings suggest that CDK4/6 inhibition combined with targeted therapies may offer an improved treatment strategy.

Targeting Polycomb Repressive Complex 2 (PRC2)

PRC2 loss through mutation of its obligate members SUZ12 or EED is common and provides a rationale for targeted combination therapies of NF1-associated tumors with bromodomain inhibitors [54, 58, 59]. The bromodomain protein BRD4 plays a crucial role in NF1-associated MPNST development and comprises a therapeutic target to potentially overcome MEKi resistance [60]. Interestingly, MPNSTs depleted of BRD4 protein exhibit a strong cytotoxic response to the pan-BET bromodomain inhibitor JQ1 [61]. Additionally, suppressing SUZ12 enhances the impact of PD-901/JQ1 administration in NF1-deficient cells [62]. In a study on NF1-mutated ovarian cancer, co-administration of JQ1 and MEKi trametinib proved effective in overcoming the common rapid drug resistance associated with single-agent MEKi [63]. A second bromodomain inhibitor, bromosporine, demonstrated a superior therapeutic index when combined with MEKi cobimetinib for treating immunotherapy-resistant NF1-mutant melanoma, compared to MEKi treatment alone [64•]. More recent work suggests DNMT1 inhibition may be a druggable dependency upon PRC2 loss, providing yet another targeted approach [65]. Overall, targeting PRC2 loss holds significant promise for enhancing existing strategies for NF1-deficient tumors by increasing cytotoxicity and limiting the development of drug resistance.

Future Therapeutic Approaches: Beyond Targeted Therapeutics

Decades of work understanding the genetic and signaling mechanisms underlying NF1-associated tumorigenesis have nominated numerous targets, yet there remains an urgent, unmet clinical need for new therapies with improved therapeutic windows and more durable responses. Promising preclinical approaches leveraging pharmacologic advances to investigate gene therapy, directly targeting Ras, or reestablishing immune system function with cellular CAR-T therapies, checkpoint inhibitors, or oncolytic viral therapy (Fig. 1B) are areas of active investigation that offer potential for the next generation of therapeutics.

Gene Therapy

Gene therapy through adeno-associated viral (AAV) vectors offers a potentially curative approach aimed at NF1 gene reconstitution. Although full-length reconstitution has been historically limited by the size of the NF1 gene, the neurofibromin GTPase-activating protein-related domain (GRD) alone, fused with an H-Ras C10 sequence, demonstrates potent ERK1/2 suppression, reduced cell growth, and exhibiting specificity for NF1-mutant MPNST cells compared to NF1-intact cells [66, 67]. However, numerous open questions remain regarding gene targeting specificity, efficient delivery, and maximum therapeutic payload size that require further research to harness the potential of neurofibromin reconstitution.

Direct Ras Inhibition

Although Ras was historically considered to be undruggable as a direct pharmacologic target, multiple covalent inhibitors targeting oncogenic Ras variants now exist. In NF-1-associated tumors lacking an oncogenic Ras variant, multiple levels of evidence support a critical role for KRAS in mediating the effects of NF1 loss [68, 69]. Accordingly, recently described pan KRAS inhibitors that inhibit wildtype KRAS yet spare NRAS and HRAS may show therapeutic efficacy for NF1 mutant tumors [70•]. However, whether KRAS is the critical Ras effector for all NF-1 manifestations remains unclear. Moreover, blocking Ras alone may be insufficient, and thus, combination approaches with existing therapies may be required to overcome resistance. Indeed, treatment resistance is a recognized problem for KRAS G12C inhibitors [71]. SHP2 inhibition has shown synergy with KRASG12C inhibitors [72, 73]. SHP2 inhibition prevents the action of SOS1/2, increasing the amount of the GDP-bound state of KRASG12C which is the target of KRAS inhibitors [74]. This is supported by the findings of KRAS-amplified cancer cell lines exhibiting increased sensitivity to SHP2 inhibition [72, 75].

CAR-T Cell Therapy

CAR-T cell therapy engineers T-cells with the ability to target overexpressed antigens specific to cancer cells and has revolutionized the treatment of cancer types, primarily hematologic malignancies such as leukemia and lymphoma [76]. Ongoing clinical trials (NCT03618381) are investigating EGFR-targeting CAR-T cell therapy for MPNSTs, and CAR-T therapy for NF1-mutated high-grade gliomas using tumor-specific internal peptides is being tested to address the challenge of non-unique expression on the surface of solid tumors. While many questions remain, including the competency of T cells derived from patients harboring a germline NF1 mutation, [77] CAR-T cell therapy is a promising area of investigation for NF1-mutant tumors.

Immune Checkpoint Inhibitors (ICI)

ICIs have revolutionized cancer care for multiple solid tumor types, and case reports suggest potential ICI efficacy in patients with MPNSTs [78]. The PD-1 inhibitor pembrolizumab was investigated in an MPMS clinical trial but was closed due to limited accrual (NCT02691026). Ongoing clinical trials are evaluating the efficacy of adjuvant nivolumab along with CTLA-4 checkpoint inhibitor ipilimumab for newly diagnosed MPNSTs (NCT04465643, NCT02834013).
Beyond PD-1 axis blockade, colony-stimulating factor-1 receptor (CSF-1R) is often upregulated in various cancer phenotypes and plays a critical role in macrophage polarization, converting tumor-associated macrophages from the tumoricidal M0 or M1 phenotype to the tumorigenic M2 phenotype [79]. Pexidartinib, a novel small molecule CSF-1R inhibitor, showed promising results in a Phase 1 study for MPNSTs when combined with sirolimus, and a Phase 2 trial is now underway (NCT02584647) [80]. MK-1775, another novel ICI, is being investigated for combating MPNSTs by inhibiting WEE1, a key regulator of cell cycle progression [81].

Oncolytic Viral (OV) Therapy

OV therapy is another promising approach for NF1-associated tumors. A measles virus-based OV approach shows efficacy in MPNST cells [82], leading to a Phase 1 trial underway to investigate the clinical efficacy of this technique (NCT02700230). Other trials leverage alternate viral agents such as Herpes Simplex Virus (HSV) HSV1716 to preferentially target actively dividing nervous system tumor cells (NCT00931931).

Conclusion

Patients with NF-1 can exhibit a diverse array of clinical manifestations. Building on classic NF1/Ras biology, MEK inhibitors are an effective therapy for a number of NF1-related manifestations, yet the heterogeneity and durability of their response motivate the development of additional approaches. Ongoing research into biologic mechanisms and signal transduction pathways dysregulated in NF1-associated tumors holds the potential to reveal additional therapeutic vulnerabilities. Moreover, targeting the tumor microenvironment and employing combination molecular therapies show promise. Continuous investigation through mechanistic investigation, preclinical modeling, clinical trials, the accumulation of long-term safety data, and collaboration between basic scientists and clinicians will be pivotal in advancing therapeutic interventions for NF1-associated tumors.

Declarations

Competing interests

The authors declare no competing interests.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
2.
Zurück zum Zitat Martin GA, et al. The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell. 1990;63:843–9.PubMedCrossRef Martin GA, et al. The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell. 1990;63:843–9.PubMedCrossRef
3.
Zurück zum Zitat Basu TN, et al. Aberrant regulation of ras proteins in malignant tumour cells from type 1 neurofibromatosis patients. Nature. 1992;356:713–5.PubMedCrossRef Basu TN, et al. Aberrant regulation of ras proteins in malignant tumour cells from type 1 neurofibromatosis patients. Nature. 1992;356:713–5.PubMedCrossRef
4.
Zurück zum Zitat DeClue JE, et al. Abnormal regulation of mammalian p21ras contributes to malignant tumor growth in von Recklinghausen (type 1) neurofibromatosis. Cell. 1992;69:265–73.PubMedCrossRef DeClue JE, et al. Abnormal regulation of mammalian p21ras contributes to malignant tumor growth in von Recklinghausen (type 1) neurofibromatosis. Cell. 1992;69:265–73.PubMedCrossRef
5.
6.
Zurück zum Zitat Gutmann DH, et al. Neurofibromatosis type 1. Nat Rev Dis Primer. 2017;3:17004.CrossRef Gutmann DH, et al. Neurofibromatosis type 1. Nat Rev Dis Primer. 2017;3:17004.CrossRef
7.
Zurück zum Zitat Spyk SL, Thomas N, Cooper DN, Upadhyaya M. Neurofibromatosis type 1-associated tumours: their somatic mutational spectrum and pathogenesis. Hum Genomics. 2011;5:623–90.PubMedCentralCrossRef Spyk SL, Thomas N, Cooper DN, Upadhyaya M. Neurofibromatosis type 1-associated tumours: their somatic mutational spectrum and pathogenesis. Hum Genomics. 2011;5:623–90.PubMedCentralCrossRef
9.
Zurück zum Zitat Zhu Y, Ghosh P, Charnay P, Burns DK, Parada LF. Neurofibromas in NF1: Schwann cell origin and role of tumor environment. Science. 2002;296:920–2.PubMedPubMedCentralCrossRef Zhu Y, Ghosh P, Charnay P, Burns DK, Parada LF. Neurofibromas in NF1: Schwann cell origin and role of tumor environment. Science. 2002;296:920–2.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Packer RJ, et al. Implications of new understandings of gliomas in children and adults with NF1: report of a consensus conference. Neuro-Oncol. 2020;22:773–84.PubMedPubMedCentralCrossRef Packer RJ, et al. Implications of new understandings of gliomas in children and adults with NF1: report of a consensus conference. Neuro-Oncol. 2020;22:773–84.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Kresbach C, et al. Atypical neurofibromas reveal distinct epigenetic features with proximity to benign peripheral nerve sheath tumor entities. Neuro-Oncol. 2023;25:1644–55.PubMedPubMedCentralCrossRef Kresbach C, et al. Atypical neurofibromas reveal distinct epigenetic features with proximity to benign peripheral nerve sheath tumor entities. Neuro-Oncol. 2023;25:1644–55.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat • Rhodes SD, et al. Cdkn2a (Arf) loss drives NF1-associated atypical neurofibroma and malignant transformation. Hum Mol Genet. 2019;28:2752–62. (This is an important study that defines the molecular mechanisms underlying malignant transformation of NF-1-associated plexiform neurofibromas to malignant peripheral nerve sheath tumors.)PubMedPubMedCentralCrossRef • Rhodes SD, et al. Cdkn2a (Arf) loss drives NF1-associated atypical neurofibroma and malignant transformation. Hum Mol Genet. 2019;28:2752–62. (This is an important study that defines the molecular mechanisms underlying malignant transformation of NF-1-associated plexiform neurofibromas to malignant peripheral nerve sheath tumors.)PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Kahen EJ, et al. Neurofibromin level directs RAS pathway signaling and mediates sensitivity to targeted agents in malignant peripheral nerve sheath tumors. Oncotarget. 2018;9:22571–85.PubMedPubMedCentralCrossRef Kahen EJ, et al. Neurofibromin level directs RAS pathway signaling and mediates sensitivity to targeted agents in malignant peripheral nerve sheath tumors. Oncotarget. 2018;9:22571–85.PubMedPubMedCentralCrossRef
17.
18.
Zurück zum Zitat • Gross AM, et al. Selumetinib in children with inoperable plexiform neurofibromas. N Engl J Med. 2020;382:1430–42. (This clinical trial evaluating the efficacy of the MEK inhibitor selumetinib led to the FDA approval of selumetinib for symptomatic, inoperable plexiform neurofibromas in NF-1 pediatric patients.)PubMedPubMedCentralCrossRef • Gross AM, et al. Selumetinib in children with inoperable plexiform neurofibromas. N Engl J Med. 2020;382:1430–42. (This clinical trial evaluating the efficacy of the MEK inhibitor selumetinib led to the FDA approval of selumetinib for symptomatic, inoperable plexiform neurofibromas in NF-1 pediatric patients.)PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat O’Sullivan Coyne GH, et al. Phase II trial of the MEK 1/2 inhibitor selumetinib (AZD6244, ARRY-142886 Hydrogen Sulfate) in adults with neurofibromatosis type 1 (NF1) and inoperable plexiform neurofibromas (PN). J Clin Oncol. 2020;38:3612–3612.CrossRef O’Sullivan Coyne GH, et al. Phase II trial of the MEK 1/2 inhibitor selumetinib (AZD6244, ARRY-142886 Hydrogen Sulfate) in adults with neurofibromatosis type 1 (NF1) and inoperable plexiform neurofibromas (PN). J Clin Oncol. 2020;38:3612–3612.CrossRef
20.
Zurück zum Zitat Bendell JC, et al. A phase 1 dose-escalation and expansion study of binimetinib (MEK162), a potent and selective oral MEK1/2 inhibitor. Br J Cancer. 2017;116:575–83.PubMedPubMedCentralCrossRef Bendell JC, et al. A phase 1 dose-escalation and expansion study of binimetinib (MEK162), a potent and selective oral MEK1/2 inhibitor. Br J Cancer. 2017;116:575–83.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Fangusaro J, et al. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial. Lancet Oncol. 2019;20:1011–22.PubMedPubMedCentralCrossRef Fangusaro J, et al. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial. Lancet Oncol. 2019;20:1011–22.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Ma Y, et al. A molecular basis for neurofibroma-associated skeletal manifestations in NF1. Genet Med Off J Am Coll Med Genet. 2020;22:1786–93. Ma Y, et al. A molecular basis for neurofibroma-associated skeletal manifestations in NF1. Genet Med Off J Am Coll Med Genet. 2020;22:1786–93.
25.
Zurück zum Zitat Ji RR, Baba H, Brenner GJ, Woolf CJ. Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat Neurosci. 1999;2:1114–9.PubMedCrossRef Ji RR, Baba H, Brenner GJ, Woolf CJ. Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat Neurosci. 1999;2:1114–9.PubMedCrossRef
26.
27.
Zurück zum Zitat Degirmenci U, Yap J, Sim YRM, Qin S, Hu J. Drug resistance in targeted cancer therapies with RAF inhibitors. Cancer Drug Resist. 2021;4:665–83.PubMedPubMedCentral Degirmenci U, Yap J, Sim YRM, Qin S, Hu J. Drug resistance in targeted cancer therapies with RAF inhibitors. Cancer Drug Resist. 2021;4:665–83.PubMedPubMedCentral
28.
Zurück zum Zitat • Kilburn LB, et al. The type II RAF inhibitor tovorafenib in relapsed/refractory pediatric low-grade glioma: the phase 2 FIREFLY-1 trial. Nat Med. 2024;30:207–17. (This is an important clinical trial demonstrating response in heavily pre-treated pediatric patients with BRAF-altered low-grade glioma that has implications for patients with NF1-associated low-grade gliomas.)PubMedCrossRef • Kilburn LB, et al. The type II RAF inhibitor tovorafenib in relapsed/refractory pediatric low-grade glioma: the phase 2 FIREFLY-1 trial. Nat Med. 2024;30:207–17. (This is an important clinical trial demonstrating response in heavily pre-treated pediatric patients with BRAF-altered low-grade glioma that has implications for patients with NF1-associated low-grade gliomas.)PubMedCrossRef
29.
Zurück zum Zitat Sigaud R, et al. The first-in-class ERK inhibitor ulixertinib shows promising activity in mitogen-activated protein kinase (MAPK)-driven pediatric low-grade glioma models. Neuro-Oncol. 2022;25:566–79.PubMedCentralCrossRef Sigaud R, et al. The first-in-class ERK inhibitor ulixertinib shows promising activity in mitogen-activated protein kinase (MAPK)-driven pediatric low-grade glioma models. Neuro-Oncol. 2022;25:566–79.PubMedCentralCrossRef
30.
Zurück zum Zitat Flint AC, et al. Combined CDK4/6 and ERK1/2 inhibition enhances antitumor activity in NF1-associated plexiform neurofibroma. Clin Cancer Res. 2023;29:3438–56.PubMedPubMedCentralCrossRef Flint AC, et al. Combined CDK4/6 and ERK1/2 inhibition enhances antitumor activity in NF1-associated plexiform neurofibroma. Clin Cancer Res. 2023;29:3438–56.PubMedPubMedCentralCrossRef
31.
32.
Zurück zum Zitat Stathis A, et al. Results of an open-label phase 1b study of the ERK inhibitor MK-8353 plus the MEK inhibitor selumetinib in patients with advanced or metastatic solid tumors. Invest New Drugs. 2023;41:1–11.CrossRef Stathis A, et al. Results of an open-label phase 1b study of the ERK inhibitor MK-8353 plus the MEK inhibitor selumetinib in patients with advanced or metastatic solid tumors. Invest New Drugs. 2023;41:1–11.CrossRef
34.
Zurück zum Zitat Yang Y, Li S, Wang Y, Zhao Y, Li Q. Protein tyrosine kinase inhibitor resistance in malignant tumors: molecular mechanisms and future perspective. Signal Transduct Target Ther. 2022;7:1–36. Yang Y, Li S, Wang Y, Zhao Y, Li Q. Protein tyrosine kinase inhibitor resistance in malignant tumors: molecular mechanisms and future perspective. Signal Transduct Target Ther. 2022;7:1–36.
35.
Zurück zum Zitat Ferguson MJ, et al. Preclinical evidence for the use of sunitinib malate in the treatment of plexiform neurofibromas. Pediatr Blood Cancer. 2016;63:206–13.PubMedCrossRef Ferguson MJ, et al. Preclinical evidence for the use of sunitinib malate in the treatment of plexiform neurofibromas. Pediatr Blood Cancer. 2016;63:206–13.PubMedCrossRef
37.
Zurück zum Zitat Robertson KA, et al. Imatinib mesylate for plexiform neurofibromas in patients with neurofibromatosis type 1: a phase 2 trial. Lancet Oncol. 2012;13:1218–24.PubMedPubMedCentralCrossRef Robertson KA, et al. Imatinib mesylate for plexiform neurofibromas in patients with neurofibromatosis type 1: a phase 2 trial. Lancet Oncol. 2012;13:1218–24.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Kim A, et al. Phase I trial and pharmacokinetic study of sorafenib in children with neurofibromatosis type I and plexiform neurofibromas. Pediatr Blood Cancer. 2013;60:396–401.PubMedCrossRef Kim A, et al. Phase I trial and pharmacokinetic study of sorafenib in children with neurofibromatosis type I and plexiform neurofibromas. Pediatr Blood Cancer. 2013;60:396–401.PubMedCrossRef
39.
Zurück zum Zitat Solares I, Viñal D, Morales-Conejo M, Rodriguez-Salas N, Feliu J. Novel molecular targeted therapies for patients with neurofibromatosis type 1 with inoperable plexiform neurofibromas: a comprehensive review. ESMO Open. 2021;6:100223.PubMedPubMedCentralCrossRef Solares I, Viñal D, Morales-Conejo M, Rodriguez-Salas N, Feliu J. Novel molecular targeted therapies for patients with neurofibromatosis type 1 with inoperable plexiform neurofibromas: a comprehensive review. ESMO Open. 2021;6:100223.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Chen Y-NP, et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 2016;535:148–52.PubMedCrossRef Chen Y-NP, et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 2016;535:148–52.PubMedCrossRef
43.
Zurück zum Zitat Wang J, et al. Combined inhibition of SHP2 and MEK is effective in models of NF1-deficient malignant peripheral nerve sheath tumors. Cancer Res. 2020;80:5367–79.PubMedPubMedCentralCrossRef Wang J, et al. Combined inhibition of SHP2 and MEK is effective in models of NF1-deficient malignant peripheral nerve sheath tumors. Cancer Res. 2020;80:5367–79.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Dasgupta B, Yi Y, Chen DY, Weber JD, Gutmann DH. Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. Cancer Res. 2005;65:2755–60.PubMedCrossRef Dasgupta B, Yi Y, Chen DY, Weber JD, Gutmann DH. Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors. Cancer Res. 2005;65:2755–60.PubMedCrossRef
46.
Zurück zum Zitat Weiss B, et al. Sirolimus for non-progressive NF1-associated plexiform neurofibromas: an NF clinical trials consortium phase II study. Pediatr Blood Cancer. 2014;61:982–6.PubMedCrossRef Weiss B, et al. Sirolimus for non-progressive NF1-associated plexiform neurofibromas: an NF clinical trials consortium phase II study. Pediatr Blood Cancer. 2014;61:982–6.PubMedCrossRef
47.
Zurück zum Zitat Weiss B, et al. Sirolimus for progressive neurofibromatosis type 1–associated plexiform neurofibromas: a Neurofibromatosis Clinical Trials Consortium phase II study. Neuro-Oncol. 2015;17:596–603.PubMedCrossRef Weiss B, et al. Sirolimus for progressive neurofibromatosis type 1–associated plexiform neurofibromas: a Neurofibromatosis Clinical Trials Consortium phase II study. Neuro-Oncol. 2015;17:596–603.PubMedCrossRef
48.
Zurück zum Zitat Zehou O, et al. Absence of efficacy of everolimus in neurofibromatosis 1-related plexiform neurofibromas: results from a phase 2a trial. J Invest Dermatol. 2019;139:718–20.PubMedCrossRef Zehou O, et al. Absence of efficacy of everolimus in neurofibromatosis 1-related plexiform neurofibromas: results from a phase 2a trial. J Invest Dermatol. 2019;139:718–20.PubMedCrossRef
49.
Zurück zum Zitat Ullrich NJ, et al. A phase II study of continuous oral mTOR inhibitor everolimus for recurrent, radiographic-progressive neurofibromatosis type 1–associated pediatric low-grade glioma: a Neurofibromatosis Clinical Trials Consortium study. Neuro-Oncol. 2020;22:1527–35.PubMedPubMedCentralCrossRef Ullrich NJ, et al. A phase II study of continuous oral mTOR inhibitor everolimus for recurrent, radiographic-progressive neurofibromatosis type 1–associated pediatric low-grade glioma: a Neurofibromatosis Clinical Trials Consortium study. Neuro-Oncol. 2020;22:1527–35.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Vasudevan HN, et al. Functional interactions between neurofibromatosis tumor suppressors underlie Schwann cell tumor de-differentiation and treatment resistance. Nat Commun. 2024;15:477.PubMedPubMedCentralCrossRef Vasudevan HN, et al. Functional interactions between neurofibromatosis tumor suppressors underlie Schwann cell tumor de-differentiation and treatment resistance. Nat Commun. 2024;15:477.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Pemov A, et al. Low mutation burden and frequent loss of CDKN2A/B and SMARCA2, but not PRC2, define premalignant neurofibromatosis type 1–associated atypical neurofibromas. Neuro-Oncol. 2019;21:981–92.PubMedPubMedCentralCrossRef Pemov A, et al. Low mutation burden and frequent loss of CDKN2A/B and SMARCA2, but not PRC2, define premalignant neurofibromatosis type 1–associated atypical neurofibromas. Neuro-Oncol. 2019;21:981–92.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Lee W, et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat Genet. 2014;46:1227–32.PubMedPubMedCentralCrossRef Lee W, et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat Genet. 2014;46:1227–32.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Williams KB, Largaespada DA. New model systems and the development of targeted therapies for the treatment of neurofibromatosis type 1-associated malignant peripheral nerve sheath tumors. Genes. 2020;11:477.PubMedPubMedCentralCrossRef Williams KB, Largaespada DA. New model systems and the development of targeted therapies for the treatment of neurofibromatosis type 1-associated malignant peripheral nerve sheath tumors. Genes. 2020;11:477.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Cristofanilli M, et al. Overall survival with palbociclib and fulvestrant in women with HR+/HER2− ABC: updated exploratory analyses of PALOMA-3, a double-blind, phase iii randomized study. Clin Cancer Res. 2022;28:3433–42.PubMedPubMedCentralCrossRef Cristofanilli M, et al. Overall survival with palbociclib and fulvestrant in women with HR+/HER2− ABC: updated exploratory analyses of PALOMA-3, a double-blind, phase iii randomized study. Clin Cancer Res. 2022;28:3433–42.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Pearson A, et al. Inactivating NF1 mutations are enriched in advanced breast cancer and contribute to endocrine therapy resistance. Clin Cancer Res. 2020;26:608–22.PubMedCrossRef Pearson A, et al. Inactivating NF1 mutations are enriched in advanced breast cancer and contribute to endocrine therapy resistance. Clin Cancer Res. 2020;26:608–22.PubMedCrossRef
58.
Zurück zum Zitat De Raedt T, et al. PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies. Nature. 2014;514:247–51.PubMedCrossRef De Raedt T, et al. PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies. Nature. 2014;514:247–51.PubMedCrossRef
60.
Zurück zum Zitat Patel AJ, et al. BET bromodomain inhibition triggers apoptosis of NF1-associated malignant peripheral nerve sheath tumors through bim induction. Cell Rep. 2014;6:81–92.PubMedCrossRef Patel AJ, et al. BET bromodomain inhibition triggers apoptosis of NF1-associated malignant peripheral nerve sheath tumors through bim induction. Cell Rep. 2014;6:81–92.PubMedCrossRef
61.
Zurück zum Zitat Cooper JM, et al. Overcoming BET inhibitor resistance in malignant peripheral nerve sheath tumors. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25:3404–16.CrossRef Cooper JM, et al. Overcoming BET inhibitor resistance in malignant peripheral nerve sheath tumors. Clin Cancer Res Off J Am Assoc Cancer Res. 2019;25:3404–16.CrossRef
62.
63.
Zurück zum Zitat Kurimchak AM, et al. Intrinsic resistance to MEK inhibition through BET protein mediated kinome reprogramming in NF1-deficient ovarian cancer. Mol Cancer Res MCR. 2019;17:1721–34.PubMedCrossRef Kurimchak AM, et al. Intrinsic resistance to MEK inhibition through BET protein mediated kinome reprogramming in NF1-deficient ovarian cancer. Mol Cancer Res MCR. 2019;17:1721–34.PubMedCrossRef
64.
Zurück zum Zitat • Dar AA, et al. Bromodomain inhibition overcomes treatment resistance in distinct molecular subtypes of melanoma. Proc Natl Acad Sci U S A. 2022;119:e2206824119. (This is an important preclinical study that demonstrates that treatment-naïve and treatment-resistant NF1-mutated melanomas respond to a combination of bromosporine and cobinimetinib, suggesting a combination of MEK and bromodomain inhibition may show efficacy in NF-1-associated tumors.)PubMedPubMedCentralCrossRef • Dar AA, et al. Bromodomain inhibition overcomes treatment resistance in distinct molecular subtypes of melanoma. Proc Natl Acad Sci U S A. 2022;119:e2206824119. (This is an important preclinical study that demonstrates that treatment-naïve and treatment-resistant NF1-mutated melanomas respond to a combination of bromosporine and cobinimetinib, suggesting a combination of MEK and bromodomain inhibition may show efficacy in NF-1-associated tumors.)PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Patel AJ, et al. PRC2-inactivating mutations in cancer enhance cytotoxic response to DNMT1-targeted therapy via enhanced viral mimicry. Cancer Discov. 2022;12:2120–39.PubMedPubMedCentralCrossRef Patel AJ, et al. PRC2-inactivating mutations in cancer enhance cytotoxic response to DNMT1-targeted therapy via enhanced viral mimicry. Cancer Discov. 2022;12:2120–39.PubMedPubMedCentralCrossRef
66.
68.
Zurück zum Zitat Dasgupta B, Li W, Perry A, Gutmann DH. Glioma formation in neurofibromatosis 1 reflects preferential activation of K-RAS in astrocytes. Cancer Res. 2005;65:236–45.PubMedCrossRef Dasgupta B, Li W, Perry A, Gutmann DH. Glioma formation in neurofibromatosis 1 reflects preferential activation of K-RAS in astrocytes. Cancer Res. 2005;65:236–45.PubMedCrossRef
69.
Zurück zum Zitat Khalaf WF, et al. K-ras is critical for modulating multiple c-kit-mediated cellular functions in wild-type and Nf1+/- mast cells. J Immunol Baltim Md. 2007;1950(178):2527–34. Khalaf WF, et al. K-ras is critical for modulating multiple c-kit-mediated cellular functions in wild-type and Nf1+/- mast cells. J Immunol Baltim Md. 2007;1950(178):2527–34.
70.
Zurück zum Zitat • Kim D, et al. Pan-KRAS inhibitor disables oncogenic signalling and tumour growth. Nature. 2023;619:160–6. (This study demonstrates that pharmacologic agents inhibiting wild-type KRAS while sparing other Ras proteins can show efficacy in tumor models.)PubMedPubMedCentralCrossRef • Kim D, et al. Pan-KRAS inhibitor disables oncogenic signalling and tumour growth. Nature. 2023;619:160–6. (This study demonstrates that pharmacologic agents inhibiting wild-type KRAS while sparing other Ras proteins can show efficacy in tumor models.)PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Li T, et al. Developing SHP2-based combination therapy for KRAS-amplified cancer. JCI Insight. 2023;8:e152714. Li T, et al. Developing SHP2-based combination therapy for KRAS-amplified cancer. JCI Insight. 2023;8:e152714.
73.
Zurück zum Zitat Hallin J, et al. The KRASG12C inhibitor, MRTX849, provides insight toward therapeutic susceptibility of KRAS mutant cancers in mouse models and patients. Cancer Discov. 2020;10:54–71.PubMedCrossRef Hallin J, et al. The KRASG12C inhibitor, MRTX849, provides insight toward therapeutic susceptibility of KRAS mutant cancers in mouse models and patients. Cancer Discov. 2020;10:54–71.PubMedCrossRef
74.
Zurück zum Zitat Fedele C, et al. SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling. J Exp Med. 2020;218:e20201414.PubMedCentralCrossRef Fedele C, et al. SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling. J Exp Med. 2020;218:e20201414.PubMedCentralCrossRef
75.
Zurück zum Zitat Drilon A, et al. SHP2 inhibition sensitizes diverse oncogene-addicted solid tumors to re-treatment with targeted therapy. Cancer Discov. 2023;13:1789–801.PubMedPubMedCentralCrossRef Drilon A, et al. SHP2 inhibition sensitizes diverse oncogene-addicted solid tumors to re-treatment with targeted therapy. Cancer Discov. 2023;13:1789–801.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–5.PubMedCrossRef June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–5.PubMedCrossRef
77.
Zurück zum Zitat Ingram DA, et al. Lymphoproliferative defects in mice lacking the expression of neurofibromin: functional and biochemical consequences ofNf1 deficiency in T-cell development and function. Blood. 2002;100:3656–62.PubMedCrossRef Ingram DA, et al. Lymphoproliferative defects in mice lacking the expression of neurofibromin: functional and biochemical consequences ofNf1 deficiency in T-cell development and function. Blood. 2002;100:3656–62.PubMedCrossRef
78.
Zurück zum Zitat Larson K, et al. Pembrolizumab achieves a complete response in an NF-1 mutated, PD-L1 positive malignant peripheral nerve sheath tumor: a case report and review of the benchmarks. J Immunother Hagerstown Md. 2022;1997(45):222–6.CrossRef Larson K, et al. Pembrolizumab achieves a complete response in an NF-1 mutated, PD-L1 positive malignant peripheral nerve sheath tumor: a case report and review of the benchmarks. J Immunother Hagerstown Md. 2022;1997(45):222–6.CrossRef
80.
Zurück zum Zitat Boal LH, et al. Pediatric PK/PD phase I trial of pexidartinib in relapsed and refractory leukemias and solid tumors including neurofibromatosis type I related plexiform neurofibromas. Clin Cancer Res Off J Am Assoc Cancer Res. 2020;26:6112–21.CrossRef Boal LH, et al. Pediatric PK/PD phase I trial of pexidartinib in relapsed and refractory leukemias and solid tumors including neurofibromatosis type I related plexiform neurofibromas. Clin Cancer Res Off J Am Assoc Cancer Res. 2020;26:6112–21.CrossRef
81.
Zurück zum Zitat Fernández-Rodríguez J, et al. A high-throughput screening platform identifies novel combination treatments for Malignant Peripheral Nerve Sheath Tumors. Mol Cancer Ther. 2022;21:1246–58.PubMedPubMedCentralCrossRef Fernández-Rodríguez J, et al. A high-throughput screening platform identifies novel combination treatments for Malignant Peripheral Nerve Sheath Tumors. Mol Cancer Ther. 2022;21:1246–58.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Deyle DR, Escobar DZ, Peng K-W, Babovic-Vuksanovic D. Oncolytic measles virus as a novel therapy for malignant peripheral nerve sheath tumors. Gene. 2015;565:140–5.PubMedCrossRef Deyle DR, Escobar DZ, Peng K-W, Babovic-Vuksanovic D. Oncolytic measles virus as a novel therapy for malignant peripheral nerve sheath tumors. Gene. 2015;565:140–5.PubMedCrossRef
Metadaten
Titel
Past, Present, and Future Therapeutic Strategies for NF-1-Associated Tumors
verfasst von
Brian Na
Shilp R. Shah
Harish N. Vasudevan
Publikationsdatum
06.05.2024
Verlag
Springer US
Erschienen in
Current Oncology Reports
Print ISSN: 1523-3790
Elektronische ISSN: 1534-6269
DOI
https://doi.org/10.1007/s11912-024-01527-4

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Das sind die führenden Symptome junger Darmkrebspatienten

Darmkrebserkrankungen in jüngeren Jahren sind ein zunehmendes Problem, das häufig längere Zeit übersehen wird, gerade weil die Patienten noch nicht alt sind. Welche Anzeichen Ärzte stutzig machen sollten, hat eine Metaanalyse herausgearbeitet.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.