Quiz-summary
0 of 30 questions completed
Questions:
- 1
- 2
- 3
- 4
- 5
- 6
- 7
- 8
- 9
- 10
- 11
- 12
- 13
- 14
- 15
- 16
- 17
- 18
- 19
- 20
- 21
- 22
- 23
- 24
- 25
- 26
- 27
- 28
- 29
- 30
Information
Premium Practice Questions
You have already completed the quiz before. Hence you can not start it again.
Quiz is loading...
You must sign in or sign up to start the quiz.
You have to finish following quiz, to start this quiz:
Results
0 of 30 questions answered correctly
Your time:
Time has elapsed
Categories
- Not categorized 0%
- 1
- 2
- 3
- 4
- 5
- 6
- 7
- 8
- 9
- 10
- 11
- 12
- 13
- 14
- 15
- 16
- 17
- 18
- 19
- 20
- 21
- 22
- 23
- 24
- 25
- 26
- 27
- 28
- 29
- 30
- Answered
- Review
-
Question 1 of 30
1. Question
Consider a patient with metastatic non-small cell lung cancer who, after initial partial response to a platinum-based chemotherapy regimen, has progressed. They are subsequently initiated on an anti-PD-1 monoclonal antibody. While initially showing a favorable response, the tumor subsequently exhibits enhanced expression of programmed death-ligand 1 (PD-L1) on both tumor cells and tumor-associated macrophages, alongside a significant increase in transforming growth factor-beta (TGF-\(\beta\)) and vascular endothelial growth factor (VEGF) levels within the tumor microenvironment. Given this complex immune-suppressive milieu, which of the following therapeutic strategies would be most likely to re-sensitize the tumor to immunotherapy and improve clinical outcomes for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immunotherapy. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on standard chemotherapy and is now receiving an anti-PD-1 antibody. Despite initial partial response, the tumor exhibits increased expression of PD-L1 on tumor cells and macrophages, along with elevated levels of TGF-\(\beta\) and VEGF within the TME. The correct approach to enhancing the anti-PD-1 response in this scenario involves addressing the immunosuppressive components of the TME that are likely contributing to resistance. TGF-\(\beta\) is a potent immunosuppressive cytokine that inhibits T cell activation and proliferation, and it also promotes the differentiation of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). VEGF, while primarily known for its role in angiogenesis, also has immunosuppressive properties, contributing to T cell dysfunction and promoting the recruitment of immunosuppressive cells. Therefore, a combination strategy that targets both PD-1/PD-L1 axis and the immunosuppressive signaling pathways mediated by TGF-\(\beta\) and VEGF would be most effective. This could involve combining the anti-PD-1 antibody with a TGF-\(\beta\) inhibitor (e.g., a TGF-\(\beta\) receptor kinase inhibitor) and/or an anti-VEGF antibody. Such a combinatorial approach aims to simultaneously unleash T cell activity by blocking immune checkpoints and to reduce the immunosuppressive milieu, thereby promoting a more favorable environment for anti-tumor immunity. The other options are less optimal. While increasing the dose of the anti-PD-1 antibody might offer some benefit, it is unlikely to overcome the profound immunosuppression mediated by high levels of TGF-\(\beta\) and VEGF. Adding a cytotoxic chemotherapy agent might induce tumor cell death, but it does not directly address the immune escape mechanisms driven by the TME. Similarly, targeting only VEGF without addressing the TGF-\(\beta\) pathway or the PD-1/PD-L1 axis would likely result in a suboptimal response, as it would not fully counteract the multifaceted immunosuppression present. The rationale for combining therapies that target distinct immunosuppressive mechanisms within the TME is a cornerstone of modern immuno-oncology research and clinical practice, particularly for overcoming resistance to single-agent immunotherapies.
Incorrect
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immunotherapy. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on standard chemotherapy and is now receiving an anti-PD-1 antibody. Despite initial partial response, the tumor exhibits increased expression of PD-L1 on tumor cells and macrophages, along with elevated levels of TGF-\(\beta\) and VEGF within the TME. The correct approach to enhancing the anti-PD-1 response in this scenario involves addressing the immunosuppressive components of the TME that are likely contributing to resistance. TGF-\(\beta\) is a potent immunosuppressive cytokine that inhibits T cell activation and proliferation, and it also promotes the differentiation of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). VEGF, while primarily known for its role in angiogenesis, also has immunosuppressive properties, contributing to T cell dysfunction and promoting the recruitment of immunosuppressive cells. Therefore, a combination strategy that targets both PD-1/PD-L1 axis and the immunosuppressive signaling pathways mediated by TGF-\(\beta\) and VEGF would be most effective. This could involve combining the anti-PD-1 antibody with a TGF-\(\beta\) inhibitor (e.g., a TGF-\(\beta\) receptor kinase inhibitor) and/or an anti-VEGF antibody. Such a combinatorial approach aims to simultaneously unleash T cell activity by blocking immune checkpoints and to reduce the immunosuppressive milieu, thereby promoting a more favorable environment for anti-tumor immunity. The other options are less optimal. While increasing the dose of the anti-PD-1 antibody might offer some benefit, it is unlikely to overcome the profound immunosuppression mediated by high levels of TGF-\(\beta\) and VEGF. Adding a cytotoxic chemotherapy agent might induce tumor cell death, but it does not directly address the immune escape mechanisms driven by the TME. Similarly, targeting only VEGF without addressing the TGF-\(\beta\) pathway or the PD-1/PD-L1 axis would likely result in a suboptimal response, as it would not fully counteract the multifaceted immunosuppression present. The rationale for combining therapies that target distinct immunosuppressive mechanisms within the TME is a cornerstone of modern immuno-oncology research and clinical practice, particularly for overcoming resistance to single-agent immunotherapies.
-
Question 2 of 30
2. Question
A patient with advanced metastatic melanoma is being considered for a combination therapy regimen at ABIM – Subspecialty in Medical Oncology University. The proposed regimen includes a PD-1 inhibitor and an anti-VEGF antibody. Considering the known effects of anti-VEGF therapy on the tumor microenvironment, which of the following is the most likely significant consequence that could synergistically enhance the anti-tumor immune response in this context?
Correct
The question probes the understanding of tumor microenvironment (TME) modulation by specific therapeutic agents, focusing on the impact of anti-angiogenic therapy on immune cell infiltration. Anti-angiogenic agents, such as VEGF inhibitors, primarily target the formation of new blood vessels within the tumor. While this can starve the tumor of nutrients and oxygen, it also has significant downstream effects on the TME. A key consequence is the normalization of the remaining tumor vasculature. This normalization process can lead to reduced interstitial fluid pressure, decreased hypoxia, and importantly, a shift in the immune cell composition. Specifically, normalization of the vasculature can facilitate the infiltration of cytotoxic T lymphocytes (CTLs) and other effector immune cells into the tumor, thereby enhancing the efficacy of immunotherapies that rely on T cell activation. Conversely, it can also lead to a decrease in immunosuppressive cells like myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) that often thrive in hypoxic and poorly vascularized environments. Therefore, the most significant consequence of anti-angiogenic therapy on the TME, in the context of potentially enhancing anti-tumor immunity, is the increased infiltration of effector immune cells.
Incorrect
The question probes the understanding of tumor microenvironment (TME) modulation by specific therapeutic agents, focusing on the impact of anti-angiogenic therapy on immune cell infiltration. Anti-angiogenic agents, such as VEGF inhibitors, primarily target the formation of new blood vessels within the tumor. While this can starve the tumor of nutrients and oxygen, it also has significant downstream effects on the TME. A key consequence is the normalization of the remaining tumor vasculature. This normalization process can lead to reduced interstitial fluid pressure, decreased hypoxia, and importantly, a shift in the immune cell composition. Specifically, normalization of the vasculature can facilitate the infiltration of cytotoxic T lymphocytes (CTLs) and other effector immune cells into the tumor, thereby enhancing the efficacy of immunotherapies that rely on T cell activation. Conversely, it can also lead to a decrease in immunosuppressive cells like myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) that often thrive in hypoxic and poorly vascularized environments. Therefore, the most significant consequence of anti-angiogenic therapy on the TME, in the context of potentially enhancing anti-tumor immunity, is the increased infiltration of effector immune cells.
-
Question 3 of 30
3. Question
A 68-year-old male with metastatic non-small cell lung cancer (NSCLC) has experienced disease progression after receiving a programmed cell death protein 1 (PD-1) inhibitor. Prior genomic profiling of his tumor revealed a KRAS G12C mutation and a tumor mutational burden (TMB) of 15 mutations per megabase. A repeat biopsy of a metastatic lesion was performed to investigate potential mechanisms of resistance. Analysis of the biopsy specimen revealed a significant increase in the density of tumor-associated macrophages (TAMs) with an M2-like phenotype, a marked expansion of regulatory T cells (Tregs) within the tumor microenvironment, and a reduction in the proportion of effector CD8+ T cells compared to initial biopsies. Additionally, there was evidence of increased expression of indoleamine 2,3-dioxygenase (IDO) by stromal cells. Given these findings, which cellular component within the tumor microenvironment is most likely contributing to the observed resistance to PD-1 inhibition and warrants further therapeutic targeting?
Correct
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immunotherapy. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on a PD-1 inhibitor. The key to answering this question lies in understanding how different cellular components within the TME can contribute to or overcome immune evasion. The tumor microenvironment is a complex ecosystem comprising cancer cells, stromal cells (fibroblasts, endothelial cells), immune cells (T cells, B cells, macrophages, myeloid-derived suppressor cells), and extracellular matrix. In the context of PD-1 blockade resistance, several mechanisms can be at play. Tumor-associated macrophages (TAMs), particularly M2-polarized TAMs, are known to promote tumor growth, angiogenesis, and immune suppression by secreting immunosuppressive cytokines like IL-10 and TGF-β, and by expressing PD-L1. They can also hinder T cell infiltration and function. Cancer-associated fibroblasts (CAFs) contribute to the physical barrier of the tumor stroma, secrete growth factors, and can also induce immune suppression. Regulatory T cells (Tregs) actively suppress anti-tumor immune responses. Considering the progression on PD-1 inhibition, the patient’s tumor likely exhibits mechanisms that bypass or counteract T cell activation. While cytotoxic T lymphocytes (CTLs) are crucial for anti-tumor immunity, their presence alone does not guarantee response if other immunosuppressive elements are dominant. Tumor cell intrinsic resistance mechanisms, such as loss of MHC class I expression or activation of alternative immune checkpoints (e.g., LAG-3, TIM-3), could also be present, but the question focuses on the TME cellular composition. The scenario highlights the need to identify the dominant immunosuppressive cellular populations within the TME that are actively hindering the anti-tumor immune response, even in the presence of PD-1 blockade. Therefore, a comprehensive assessment of the TME would prioritize identifying the cellular components that are most actively contributing to immune suppression and resistance. The correct approach involves identifying the cellular component that is most strongly associated with immune suppression and resistance to PD-1 blockade, based on established knowledge of tumor immunology. This involves recognizing the multifaceted role of various immune cells and stromal components in shaping the anti-tumor immune response.
Incorrect
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immunotherapy. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on a PD-1 inhibitor. The key to answering this question lies in understanding how different cellular components within the TME can contribute to or overcome immune evasion. The tumor microenvironment is a complex ecosystem comprising cancer cells, stromal cells (fibroblasts, endothelial cells), immune cells (T cells, B cells, macrophages, myeloid-derived suppressor cells), and extracellular matrix. In the context of PD-1 blockade resistance, several mechanisms can be at play. Tumor-associated macrophages (TAMs), particularly M2-polarized TAMs, are known to promote tumor growth, angiogenesis, and immune suppression by secreting immunosuppressive cytokines like IL-10 and TGF-β, and by expressing PD-L1. They can also hinder T cell infiltration and function. Cancer-associated fibroblasts (CAFs) contribute to the physical barrier of the tumor stroma, secrete growth factors, and can also induce immune suppression. Regulatory T cells (Tregs) actively suppress anti-tumor immune responses. Considering the progression on PD-1 inhibition, the patient’s tumor likely exhibits mechanisms that bypass or counteract T cell activation. While cytotoxic T lymphocytes (CTLs) are crucial for anti-tumor immunity, their presence alone does not guarantee response if other immunosuppressive elements are dominant. Tumor cell intrinsic resistance mechanisms, such as loss of MHC class I expression or activation of alternative immune checkpoints (e.g., LAG-3, TIM-3), could also be present, but the question focuses on the TME cellular composition. The scenario highlights the need to identify the dominant immunosuppressive cellular populations within the TME that are actively hindering the anti-tumor immune response, even in the presence of PD-1 blockade. Therefore, a comprehensive assessment of the TME would prioritize identifying the cellular components that are most actively contributing to immune suppression and resistance. The correct approach involves identifying the cellular component that is most strongly associated with immune suppression and resistance to PD-1 blockade, based on established knowledge of tumor immunology. This involves recognizing the multifaceted role of various immune cells and stromal components in shaping the anti-tumor immune response.
-
Question 4 of 30
4. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Molecular profiling of his tumor tissue reveals an *EGFR* exon 19 deletion and a *KRAS* G12C mutation. He previously received a platinum-based doublet chemotherapy regimen followed by a PD-1 inhibitor, with documented disease progression after 6 months of treatment. Considering the molecular findings and treatment history, what is the most appropriate subsequent therapeutic strategy for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The tumor harbors an *EGFR* exon 19 deletion and a *KRAS* G12C mutation. The question asks for the most appropriate next line of therapy. Given the presence of an *EGFR* exon 19 deletion, an EGFR tyrosine kinase inhibitor (TKI) is indicated. Osimertinib is a third-generation EGFR TKI that is highly effective against common EGFR mutations, including exon 19 deletions, and has demonstrated superior efficacy and CNS penetration compared to earlier generation TKIs. While the *KRAS* G12C mutation is also present, targeted therapies for *KRAS* G12C (like sotorasib or adagrasib) are typically considered in patients who have progressed on or are intolerant to EGFR TKIs, or in the absence of other actionable driver mutations. In this specific context, the *EGFR* mutation is considered the primary driver and dictates the initial targeted therapy. Combination therapy with an EGFR TKI and a KRAS G12C inhibitor is still under investigation and not yet standard of care for initial treatment in this setting. Continuation of chemotherapy would be less effective than targeted therapy given the identified driver mutation. Re-challenging with immunotherapy alone is unlikely to be effective after progression on prior immunotherapy. Therefore, initiating osimertinib is the most evidence-based and appropriate next step.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The tumor harbors an *EGFR* exon 19 deletion and a *KRAS* G12C mutation. The question asks for the most appropriate next line of therapy. Given the presence of an *EGFR* exon 19 deletion, an EGFR tyrosine kinase inhibitor (TKI) is indicated. Osimertinib is a third-generation EGFR TKI that is highly effective against common EGFR mutations, including exon 19 deletions, and has demonstrated superior efficacy and CNS penetration compared to earlier generation TKIs. While the *KRAS* G12C mutation is also present, targeted therapies for *KRAS* G12C (like sotorasib or adagrasib) are typically considered in patients who have progressed on or are intolerant to EGFR TKIs, or in the absence of other actionable driver mutations. In this specific context, the *EGFR* mutation is considered the primary driver and dictates the initial targeted therapy. Combination therapy with an EGFR TKI and a KRAS G12C inhibitor is still under investigation and not yet standard of care for initial treatment in this setting. Continuation of chemotherapy would be less effective than targeted therapy given the identified driver mutation. Re-challenging with immunotherapy alone is unlikely to be effective after progression on prior immunotherapy. Therefore, initiating osimertinib is the most evidence-based and appropriate next step.
-
Question 5 of 30
5. Question
A 68-year-old male presents with a new diagnosis of metastatic non-small cell lung cancer (NSCLC). Following initial treatment with a platinum-based chemotherapy regimen and an immune checkpoint inhibitor, he experiences documented disease progression. Comprehensive genomic profiling of his tumor reveals an activating *KRAS* G12C mutation. Considering the patient’s history and molecular findings, which of the following therapeutic strategies would represent the most targeted and evidence-based next-line treatment at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the *KRAS* gene, specifically a G12C substitution. This mutation is a common driver in a subset of NSCLC. The question asks about the most appropriate next-line therapy. First, let’s establish the context. The patient has received prior platinum-based chemotherapy and an immune checkpoint inhibitor, indicating a need for a treatment that targets a specific molecular alteration or offers a different mechanism of action. The presence of a *KRAS* G12C mutation is a critical piece of information. Recent advancements in targeted therapy have led to the development of specific inhibitors for this particular mutation. Sotorasib and adagrasib are examples of such inhibitors that directly target the mutated KRAS G12C protein, inducing its inactivation and thereby inhibiting downstream signaling pathways that promote tumor growth and survival. These agents have demonstrated significant clinical activity in patients with *KRAS* G12C-mutated NSCLC who have progressed on prior therapy. Considering the available treatment options for a patient with documented *KRAS* G12C-mutated NSCLC after progression on standard first-line treatment, a KRAS G12C inhibitor represents the most targeted and evidence-based approach. Other options, such as docetaxel, are cytotoxic chemotherapy agents that may offer some benefit but lack the specificity and potentially improved toxicity profile of targeted therapy. Continuation of immunotherapy would be less likely to be effective given prior progression on such therapy, and there are no specific biomarkers mentioned that would strongly support its re-challenge or combination with another agent in this context. Angiogenesis inhibitors like bevacizumab might be considered in certain NSCLC subtypes or combinations, but a direct KRAS G12C inhibitor is the most precise therapy for this specific molecular profile. Therefore, the selection of a KRAS G12C inhibitor is the most appropriate next step in management for this patient.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the *KRAS* gene, specifically a G12C substitution. This mutation is a common driver in a subset of NSCLC. The question asks about the most appropriate next-line therapy. First, let’s establish the context. The patient has received prior platinum-based chemotherapy and an immune checkpoint inhibitor, indicating a need for a treatment that targets a specific molecular alteration or offers a different mechanism of action. The presence of a *KRAS* G12C mutation is a critical piece of information. Recent advancements in targeted therapy have led to the development of specific inhibitors for this particular mutation. Sotorasib and adagrasib are examples of such inhibitors that directly target the mutated KRAS G12C protein, inducing its inactivation and thereby inhibiting downstream signaling pathways that promote tumor growth and survival. These agents have demonstrated significant clinical activity in patients with *KRAS* G12C-mutated NSCLC who have progressed on prior therapy. Considering the available treatment options for a patient with documented *KRAS* G12C-mutated NSCLC after progression on standard first-line treatment, a KRAS G12C inhibitor represents the most targeted and evidence-based approach. Other options, such as docetaxel, are cytotoxic chemotherapy agents that may offer some benefit but lack the specificity and potentially improved toxicity profile of targeted therapy. Continuation of immunotherapy would be less likely to be effective given prior progression on such therapy, and there are no specific biomarkers mentioned that would strongly support its re-challenge or combination with another agent in this context. Angiogenesis inhibitors like bevacizumab might be considered in certain NSCLC subtypes or combinations, but a direct KRAS G12C inhibitor is the most precise therapy for this specific molecular profile. Therefore, the selection of a KRAS G12C inhibitor is the most appropriate next step in management for this patient.
-
Question 6 of 30
6. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Initial molecular profiling reveals an activating KRAS G12C mutation and wild-type p53. He received first-line therapy with pembrolizumab and platinum-based chemotherapy, achieving a partial response, but subsequently progressed after 8 months. Considering the molecular findings and the patient’s treatment history, which of the following therapeutic strategies would be most aligned with current advancements in precision oncology as taught at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The tumor harbors an activating mutation in the KRAS gene (specifically, G12C) and a wild-type p53. The question asks about the most appropriate next therapeutic step, considering emerging targeted therapies. Sotorasib is a specific inhibitor of KRAS G12C, designed to target this particular oncogenic driver mutation. While other options represent valid treatment modalities in oncology, they are not the most precise or evidence-based choice given the specific molecular profile of the tumor. For instance, continuing immunotherapy might be considered in certain contexts, but the presence of a targetable mutation often warrants its use. Docetaxel is a standard chemotherapy agent, but targeted therapy is generally preferred when a specific actionable mutation is identified. Combination chemotherapy with pemetrexed and carboplatin is a first-line regimen, not typically a subsequent line of therapy after progression on similar agents, especially when a targeted therapy is available. Therefore, the identification of the KRAS G12C mutation strongly directs the treatment towards a KRAS G12C inhibitor like sotorasib, representing a paradigm shift towards precision medicine in NSCLC. This approach aligns with the educational philosophy of ABIM – Subspecialty in Medical Oncology University, which emphasizes the integration of molecular diagnostics and targeted therapeutics in patient care.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The tumor harbors an activating mutation in the KRAS gene (specifically, G12C) and a wild-type p53. The question asks about the most appropriate next therapeutic step, considering emerging targeted therapies. Sotorasib is a specific inhibitor of KRAS G12C, designed to target this particular oncogenic driver mutation. While other options represent valid treatment modalities in oncology, they are not the most precise or evidence-based choice given the specific molecular profile of the tumor. For instance, continuing immunotherapy might be considered in certain contexts, but the presence of a targetable mutation often warrants its use. Docetaxel is a standard chemotherapy agent, but targeted therapy is generally preferred when a specific actionable mutation is identified. Combination chemotherapy with pemetrexed and carboplatin is a first-line regimen, not typically a subsequent line of therapy after progression on similar agents, especially when a targeted therapy is available. Therefore, the identification of the KRAS G12C mutation strongly directs the treatment towards a KRAS G12C inhibitor like sotorasib, representing a paradigm shift towards precision medicine in NSCLC. This approach aligns with the educational philosophy of ABIM – Subspecialty in Medical Oncology University, which emphasizes the integration of molecular diagnostics and targeted therapeutics in patient care.
-
Question 7 of 30
7. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Molecular profiling of his tumor reveals an activating KRAS G12C mutation and a programmed death-ligand 1 (PD-L1) tumor proportion score (TPS) of 60%. He initially received first-line therapy with pembrolizumab and platinum-doublet chemotherapy, achieving a partial response. However, after 8 months of treatment, he experienced documented disease progression. Considering the available therapeutic options and the patient’s molecular profile, what is the most appropriate subsequent treatment strategy for this individual at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C, and exhibits a PD-L1 TPS of 60%. The question asks for the most appropriate next line of therapy. Given the progression on prior immunotherapy, re-challenging with a PD-1 inhibitor alone is unlikely to be effective. While chemotherapy remains an option, targeted therapy directed at the KRAS G12C mutation is now a standard of care for patients with this specific alteration. Sotorasib and adagrasib are approved KRAS G12C inhibitors. The efficacy of these agents is well-established in this patient population, offering a more personalized and potentially less toxic approach than broad-spectrum chemotherapy. Combination therapy with chemotherapy and immunotherapy might be considered in certain contexts, but given the specific actionable mutation and prior progression on immunotherapy, targeting the KRAS G12C mutation directly is the most rational and evidence-based next step. Therefore, initiating treatment with a KRAS G12C inhibitor is the preferred strategy.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C, and exhibits a PD-L1 TPS of 60%. The question asks for the most appropriate next line of therapy. Given the progression on prior immunotherapy, re-challenging with a PD-1 inhibitor alone is unlikely to be effective. While chemotherapy remains an option, targeted therapy directed at the KRAS G12C mutation is now a standard of care for patients with this specific alteration. Sotorasib and adagrasib are approved KRAS G12C inhibitors. The efficacy of these agents is well-established in this patient population, offering a more personalized and potentially less toxic approach than broad-spectrum chemotherapy. Combination therapy with chemotherapy and immunotherapy might be considered in certain contexts, but given the specific actionable mutation and prior progression on immunotherapy, targeting the KRAS G12C mutation directly is the most rational and evidence-based next step. Therefore, initiating treatment with a KRAS G12C inhibitor is the preferred strategy.
-
Question 8 of 30
8. Question
A 68-year-old male presents with a new diagnosis of metastatic non-small cell lung cancer (NSCLC), adenocarcinoma histology. He received initial treatment with pembrolizumab and platinum-doublet chemotherapy, achieving a partial response. However, after 8 months of therapy, surveillance imaging reveals progressive disease. Comprehensive genomic profiling of his tumor specimen identifies a KRAS G12C mutation, with no other actionable driver mutations detected. Considering the patient’s history of progression on immunotherapy and the presence of this specific molecular alteration, which of the following therapeutic strategies would represent the most evidence-based next step in management for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation. The question asks for the most appropriate next line of therapy. Current guidelines and clinical trial data support the use of KRAS G12C inhibitors, such as sotorasib or adagrasib, in patients with this specific mutation who have progressed on prior treatments. These targeted therapies directly inhibit the mutated KRAS protein, which is a key driver of tumor growth in this subset of NSCLC. The efficacy of these agents has been demonstrated in clinical trials, showing meaningful objective response rates and progression-free survival. Therefore, the correct approach involves identifying the specific molecular alteration driving the cancer and selecting a therapy that targets that alteration. In this case, the KRAS G12C mutation is actionable with available targeted agents. Other options, such as continuing broad-spectrum chemotherapy or switching to a different class of immunotherapy without a clear predictive biomarker for that specific agent, are less likely to be as effective given the prior progression on immunotherapy and the presence of a targetable mutation. While a biopsy for further molecular profiling is always a consideration in evolving treatment landscapes, the immediate availability of a KRAS G12C inhibitor makes it the most logical and evidence-based next step.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation. The question asks for the most appropriate next line of therapy. Current guidelines and clinical trial data support the use of KRAS G12C inhibitors, such as sotorasib or adagrasib, in patients with this specific mutation who have progressed on prior treatments. These targeted therapies directly inhibit the mutated KRAS protein, which is a key driver of tumor growth in this subset of NSCLC. The efficacy of these agents has been demonstrated in clinical trials, showing meaningful objective response rates and progression-free survival. Therefore, the correct approach involves identifying the specific molecular alteration driving the cancer and selecting a therapy that targets that alteration. In this case, the KRAS G12C mutation is actionable with available targeted agents. Other options, such as continuing broad-spectrum chemotherapy or switching to a different class of immunotherapy without a clear predictive biomarker for that specific agent, are less likely to be as effective given the prior progression on immunotherapy and the presence of a targetable mutation. While a biopsy for further molecular profiling is always a consideration in evolving treatment landscapes, the immediate availability of a KRAS G12C inhibitor makes it the most logical and evidence-based next step.
-
Question 9 of 30
9. Question
A 62-year-old male with advanced EGFR-mutated non-small cell lung cancer (NSCLC) initially responded well to gefitinib. After 18 months, he presents with progressive disease. Molecular profiling of his tumor biopsy reveals a new point mutation in the EGFR gene that significantly alters the allosteric binding site of the drug. Considering the known mechanisms of acquired resistance to EGFR tyrosine kinase inhibitors, which of the following represents the most likely molecular alteration driving this observed clinical progression and necessitating a change in therapeutic approach at ABIM – Subspecialty in Medical Oncology University?
Correct
The question probes the understanding of mechanisms of resistance to targeted therapies, specifically focusing on the role of acquired mutations in receptor tyrosine kinases. In the context of non-small cell lung cancer (NSCLC) treated with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), the development of resistance is a critical clinical challenge. While several mechanisms exist, including bypass signaling pathways and amplification of oncogenic drivers, the acquisition of secondary mutations within the target receptor itself is a well-established and clinically significant cause of TKI failure. Specifically, the T790M mutation in EGFR confers resistance to first- and second-generation EGFR TKIs by altering the binding pocket and reducing drug affinity. This acquired resistance mechanism necessitates a change in therapeutic strategy, often involving third-generation TKIs that are designed to overcome this specific mutation. Therefore, identifying the presence of the T790M mutation is paramount for guiding subsequent treatment decisions in patients who have progressed on earlier-generation EGFR TKIs. The other options represent mechanisms of resistance that are either less common, not directly related to acquired mutations in the primary target, or represent a different class of resistance altogether. For instance, amplification of MET can lead to resistance through bypass signaling, but it is not a direct mutation within the EGFR itself. Activation of alternative signaling pathways like PI3K/AKT or MAPK can also contribute to resistance, but the question specifically asks about a mechanism directly related to the target receptor’s altered function due to a new mutation. Upregulation of drug efflux pumps, such as P-glycoprotein, is a known mechanism of multidrug resistance but is not the primary driver of resistance to EGFR TKIs in the context of acquired mutations within the receptor.
Incorrect
The question probes the understanding of mechanisms of resistance to targeted therapies, specifically focusing on the role of acquired mutations in receptor tyrosine kinases. In the context of non-small cell lung cancer (NSCLC) treated with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), the development of resistance is a critical clinical challenge. While several mechanisms exist, including bypass signaling pathways and amplification of oncogenic drivers, the acquisition of secondary mutations within the target receptor itself is a well-established and clinically significant cause of TKI failure. Specifically, the T790M mutation in EGFR confers resistance to first- and second-generation EGFR TKIs by altering the binding pocket and reducing drug affinity. This acquired resistance mechanism necessitates a change in therapeutic strategy, often involving third-generation TKIs that are designed to overcome this specific mutation. Therefore, identifying the presence of the T790M mutation is paramount for guiding subsequent treatment decisions in patients who have progressed on earlier-generation EGFR TKIs. The other options represent mechanisms of resistance that are either less common, not directly related to acquired mutations in the primary target, or represent a different class of resistance altogether. For instance, amplification of MET can lead to resistance through bypass signaling, but it is not a direct mutation within the EGFR itself. Activation of alternative signaling pathways like PI3K/AKT or MAPK can also contribute to resistance, but the question specifically asks about a mechanism directly related to the target receptor’s altered function due to a new mutation. Upregulation of drug efflux pumps, such as P-glycoprotein, is a known mechanism of multidrug resistance but is not the primary driver of resistance to EGFR TKIs in the context of acquired mutations within the receptor.
-
Question 10 of 30
10. Question
A 68-year-old gentleman presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Initial treatment with pembrolizumab and carboplatin/pemetrexed resulted in a partial response, but after 10 months, radiographic progression is evident. Comprehensive genomic profiling of the tumor tissue reveals a KRAS G12C mutation. The patient has no contraindications to further therapy and is eager to pursue treatment options that leverage molecular insights. Considering the principles of precision medicine emphasized at ABIM – Subspecialty in Medical Oncology University, which of the following therapeutic strategies would represent the most appropriate next step in management?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. The question asks for the most appropriate next therapeutic step. Given the documented KRAS G12C mutation, a targeted therapy specifically designed to inhibit this mutated protein is the most logical and evidence-based approach. Sotorasib and adagrasib are approved KRAS G12C inhibitors. These agents work by binding to the mutated KRAS protein in its inactive GTP-bound state, preventing downstream signaling pathways that promote cell proliferation and survival. This targeted approach offers a higher likelihood of response and potentially better tolerability compared to broad-spectrum cytotoxic chemotherapy or further non-specific immunotherapy in this context. While other options might be considered in different clinical scenarios (e.g., docetaxel for second-line chemotherapy, nivolumab if the patient had not received prior immunotherapy or had a different response profile, or a different targeted agent if a different mutation was present), the presence of a targetable KRAS G12C mutation makes a specific inhibitor the preferred next step for patients enrolled in advanced medical oncology training programs at institutions like ABIM – Subspecialty in Medical Oncology University.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. The question asks for the most appropriate next therapeutic step. Given the documented KRAS G12C mutation, a targeted therapy specifically designed to inhibit this mutated protein is the most logical and evidence-based approach. Sotorasib and adagrasib are approved KRAS G12C inhibitors. These agents work by binding to the mutated KRAS protein in its inactive GTP-bound state, preventing downstream signaling pathways that promote cell proliferation and survival. This targeted approach offers a higher likelihood of response and potentially better tolerability compared to broad-spectrum cytotoxic chemotherapy or further non-specific immunotherapy in this context. While other options might be considered in different clinical scenarios (e.g., docetaxel for second-line chemotherapy, nivolumab if the patient had not received prior immunotherapy or had a different response profile, or a different targeted agent if a different mutation was present), the presence of a targetable KRAS G12C mutation makes a specific inhibitor the preferred next step for patients enrolled in advanced medical oncology training programs at institutions like ABIM – Subspecialty in Medical Oncology University.
-
Question 11 of 30
11. Question
A 62-year-old male with a history of heavy smoking presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Immunohistochemical staining of his tumor biopsy reveals high PD-L1 expression (TPS 80%), a significant infiltrate of CD8+ cytotoxic T lymphocytes within the tumor stroma, and marked stromal desmoplasia with abundant cancer-associated fibroblasts (CAFs). He was initiated on pembrolizumab monotherapy but experienced progressive disease after three cycles, evidenced by new metastatic lesions on imaging. Considering the tumor microenvironment characteristics and the observed treatment failure, which of the following therapeutic strategies would be most likely to re-sensitize his tumor to immune checkpoint inhibition or provide a synergistic anti-tumor effect?
Correct
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immunotherapy. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on a PD-1 inhibitor. The tumor exhibits high expression of PD-L1, a CD8+ T cell infiltrate, and significant stromal desmoplasia with abundant cancer-associated fibroblasts (CAFs). The core concept being tested is how different components of the TME can either synergize with or antagonize the effects of immunotherapy. While PD-L1 expression and CD8+ T cell infiltration are generally considered favorable prognostic markers and indicators of potential response to PD-1/PD-L1 blockade, the presence of extensive desmoplasia and CAFs can create a physical barrier to T cell infiltration and function. CAFs can also secrete immunosuppressive factors, remodel the extracellular matrix (ECM) in ways that hinder immune cell trafficking, and promote angiogenesis that is not conducive to anti-tumor immunity. In this context, the patient’s progression despite PD-L1 positivity and CD8+ infiltration suggests that other TME factors are overriding the potential benefits of checkpoint blockade. The dense stromal component, characterized by desmoplasia and CAFs, is a well-established mechanism of resistance to immunotherapy. These fibroblasts can impede T cell penetration into the tumor core, reduce the availability of effector cytokines, and promote an immunosuppressive milieu. Therefore, targeting these stromal elements or their products is a logical next step to overcome resistance. Option a) focuses on directly targeting the fibroblasts and their matrix-remodeling capabilities. Inhibiting CAFs or their secreted factors, such as collagen or growth factors that promote desmoplasia, could potentially normalize the TME, improve T cell infiltration, and restore sensitivity to PD-1 blockade or other immunotherapies. This approach directly addresses the identified resistance mechanism. Option b) suggests increasing the dose of the PD-1 inhibitor. While dose escalation can sometimes be considered, the patient has already progressed on the standard dose, and the underlying issue appears to be TME-mediated resistance rather than insufficient target engagement. Simply increasing the dose is unlikely to overcome a physical barrier or a profoundly immunosuppressive stromal environment. Option c) proposes adding a VEGF inhibitor. While VEGF plays a role in angiogenesis and can influence immune cell infiltration, its primary role is in promoting abnormal tumor vasculature. While anti-angiogenic therapy can sometimes synergize with immunotherapy, the prominent feature described is desmoplasia and CAF activity, which are not solely dependent on VEGF. Targeting VEGF might not be as directly impactful as addressing the fibrotic stroma itself. Option d) suggests switching to a different cytotoxic chemotherapy. While chemotherapy can have immunomodulatory effects, it does not directly address the fibrotic TME that is likely causing resistance to the current immunotherapy. A switch to chemotherapy without modulating the TME might not overcome the fundamental barrier to immune recognition and killing. Therefore, the most rational approach to overcome resistance in this scenario, given the prominent desmoplasia and CAF presence, is to target the fibrotic stroma and its associated cellular components.
Incorrect
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immunotherapy. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on a PD-1 inhibitor. The tumor exhibits high expression of PD-L1, a CD8+ T cell infiltrate, and significant stromal desmoplasia with abundant cancer-associated fibroblasts (CAFs). The core concept being tested is how different components of the TME can either synergize with or antagonize the effects of immunotherapy. While PD-L1 expression and CD8+ T cell infiltration are generally considered favorable prognostic markers and indicators of potential response to PD-1/PD-L1 blockade, the presence of extensive desmoplasia and CAFs can create a physical barrier to T cell infiltration and function. CAFs can also secrete immunosuppressive factors, remodel the extracellular matrix (ECM) in ways that hinder immune cell trafficking, and promote angiogenesis that is not conducive to anti-tumor immunity. In this context, the patient’s progression despite PD-L1 positivity and CD8+ infiltration suggests that other TME factors are overriding the potential benefits of checkpoint blockade. The dense stromal component, characterized by desmoplasia and CAFs, is a well-established mechanism of resistance to immunotherapy. These fibroblasts can impede T cell penetration into the tumor core, reduce the availability of effector cytokines, and promote an immunosuppressive milieu. Therefore, targeting these stromal elements or their products is a logical next step to overcome resistance. Option a) focuses on directly targeting the fibroblasts and their matrix-remodeling capabilities. Inhibiting CAFs or their secreted factors, such as collagen or growth factors that promote desmoplasia, could potentially normalize the TME, improve T cell infiltration, and restore sensitivity to PD-1 blockade or other immunotherapies. This approach directly addresses the identified resistance mechanism. Option b) suggests increasing the dose of the PD-1 inhibitor. While dose escalation can sometimes be considered, the patient has already progressed on the standard dose, and the underlying issue appears to be TME-mediated resistance rather than insufficient target engagement. Simply increasing the dose is unlikely to overcome a physical barrier or a profoundly immunosuppressive stromal environment. Option c) proposes adding a VEGF inhibitor. While VEGF plays a role in angiogenesis and can influence immune cell infiltration, its primary role is in promoting abnormal tumor vasculature. While anti-angiogenic therapy can sometimes synergize with immunotherapy, the prominent feature described is desmoplasia and CAF activity, which are not solely dependent on VEGF. Targeting VEGF might not be as directly impactful as addressing the fibrotic stroma itself. Option d) suggests switching to a different cytotoxic chemotherapy. While chemotherapy can have immunomodulatory effects, it does not directly address the fibrotic TME that is likely causing resistance to the current immunotherapy. A switch to chemotherapy without modulating the TME might not overcome the fundamental barrier to immune recognition and killing. Therefore, the most rational approach to overcome resistance in this scenario, given the prominent desmoplasia and CAF presence, is to target the fibrotic stroma and its associated cellular components.
-
Question 12 of 30
12. Question
A 68-year-old male presents with a persistent cough and unintended weight loss. Imaging reveals metastatic non-small cell lung cancer (NSCLC) involving the liver and adrenal glands. Comprehensive genomic profiling of the tumor tissue demonstrates an *EGFR* exon 19 deletion and a *KRAS* G12C mutation. The patient previously received first-line treatment with carboplatin, paclitaxel, and pembrolizumab, after which he experienced disease progression. Considering the molecular findings and prior treatment, what is the most appropriate subsequent therapeutic strategy for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and pembrolizumab. The patient’s tumor harbors an activating mutation in the *EGFR* gene (specifically, an exon 19 deletion) and a *KRAS* G12C mutation. The question asks for the most appropriate next line of therapy. First, consider the *EGFR* mutation. Activating *EGFR* mutations are typically treated with tyrosine kinase inhibitors (TKIs). The presence of an *EGFR* exon 19 deletion is a well-established predictive biomarker for response to *EGFR* TKIs. Therefore, initiating an *EGFR* TKI is a primary consideration. Next, consider the *KRAS* G12C mutation. While *KRAS* mutations are common drivers in NSCLC, they were historically considered “undruggable.” However, the development of specific inhibitors targeting the G12C mutation has changed this landscape. Sotorasib and adagrasib are examples of such inhibitors. The critical decision point arises when a patient has multiple oncogenic driver mutations. In NSCLC, the general principle is to target the most actionable mutation, and *EGFR* mutations are highly actionable with potent TKIs. However, the presence of a co-occurring *KRAS* G12C mutation complicates this. Current understanding and clinical practice guidelines suggest that if an *EGFR* activating mutation is present, it should be targeted first, as *EGFR* TKIs have demonstrated significant efficacy in this setting. While *KRAS* G12C inhibitors are also effective, their role in the context of concurrent *EGFR* mutations is less established, and targeting the *EGFR* mutation first is the standard approach. Furthermore, the question implies progression after a platinum-based chemotherapy and pembrolizumab, meaning the patient has already received first-line treatment. Therefore, the most appropriate next step is to target the *EGFR* mutation with an *EGFR* TKI. Among the options, an *EGFR* TKI is the most suitable choice given the identified *EGFR* exon 19 deletion. The *KRAS* G12C mutation, while present, would typically be addressed in a subsequent line of therapy if the *EGFR* TKI fails, or in specific clinical trial settings, but not as the primary initial treatment when a highly actionable *EGFR* mutation is present.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and pembrolizumab. The patient’s tumor harbors an activating mutation in the *EGFR* gene (specifically, an exon 19 deletion) and a *KRAS* G12C mutation. The question asks for the most appropriate next line of therapy. First, consider the *EGFR* mutation. Activating *EGFR* mutations are typically treated with tyrosine kinase inhibitors (TKIs). The presence of an *EGFR* exon 19 deletion is a well-established predictive biomarker for response to *EGFR* TKIs. Therefore, initiating an *EGFR* TKI is a primary consideration. Next, consider the *KRAS* G12C mutation. While *KRAS* mutations are common drivers in NSCLC, they were historically considered “undruggable.” However, the development of specific inhibitors targeting the G12C mutation has changed this landscape. Sotorasib and adagrasib are examples of such inhibitors. The critical decision point arises when a patient has multiple oncogenic driver mutations. In NSCLC, the general principle is to target the most actionable mutation, and *EGFR* mutations are highly actionable with potent TKIs. However, the presence of a co-occurring *KRAS* G12C mutation complicates this. Current understanding and clinical practice guidelines suggest that if an *EGFR* activating mutation is present, it should be targeted first, as *EGFR* TKIs have demonstrated significant efficacy in this setting. While *KRAS* G12C inhibitors are also effective, their role in the context of concurrent *EGFR* mutations is less established, and targeting the *EGFR* mutation first is the standard approach. Furthermore, the question implies progression after a platinum-based chemotherapy and pembrolizumab, meaning the patient has already received first-line treatment. Therefore, the most appropriate next step is to target the *EGFR* mutation with an *EGFR* TKI. Among the options, an *EGFR* TKI is the most suitable choice given the identified *EGFR* exon 19 deletion. The *KRAS* G12C mutation, while present, would typically be addressed in a subsequent line of therapy if the *EGFR* TKI fails, or in specific clinical trial settings, but not as the primary initial treatment when a highly actionable *EGFR* mutation is present.
-
Question 13 of 30
13. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Initial molecular profiling reveals an *EGFR* exon 19 deletion and a *KRAS* G12C mutation. He receives standard platinum-doublet chemotherapy concurrently with pembrolizumab, achieving a partial response. However, after 6 months of therapy, he develops progressive disease. A repeat biopsy confirms the presence of the *EGFR* exon 19 deletion and the *KRAS* G12C mutation, with no new significant mutations identified. Considering the established treatment paradigms at ABIM – Subspecialty in Medical Oncology University, what is the most appropriate next therapeutic intervention for this patient?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and pembrolizumab. The patient’s tumor harbors an activating mutation in the *EGFR* gene (specifically, an exon 19 deletion) and a *KRAS* G12C mutation. The question asks about the most appropriate next step in management. First, it’s crucial to recognize the presence of actionable driver mutations in NSCLC. *EGFR* mutations, such as exon 19 deletions or L858R point mutations, are well-established targets for tyrosine kinase inhibitors (TKIs). Similarly, *KRAS* mutations, particularly the G12C variant, have become targetable with the development of specific inhibitors. In a patient who has progressed on immunotherapy and chemotherapy, re-biopsy and molecular profiling are often indicated to identify new or persistent actionable mutations that can guide subsequent therapy. Given the documented *EGFR* exon 19 deletion, treatment with an *EGFR* TKI is the standard of care for such patients, as these agents have demonstrated superior efficacy and tolerability compared to chemotherapy in this setting. Osimertinib, a third-generation *EGFR* TKI, is particularly effective against both sensitizing *EGFR* mutations and the T790M resistance mutation, and it also has activity against *KRAS* mutations. While the patient also has a *KRAS* G12C mutation, which is also targetable, the *EGFR* mutation is generally considered the primary driver in this context, and *EGFR* TKIs are typically prioritized when both are present, especially after progression on immunotherapy. The development of resistance to prior treatments, including immunotherapy, necessitates a re-evaluation of the tumor’s molecular landscape. Therefore, initiating therapy with an *EGFR* TKI like osimertinib is the most evidence-based and effective approach to re-challenge the tumor and potentially achieve disease control. Other options, such as continuing chemotherapy or immunotherapy, are less likely to be effective given the documented progression on these modalities. Investigating other less common mutations without addressing the known driver mutations would be a suboptimal strategy.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and pembrolizumab. The patient’s tumor harbors an activating mutation in the *EGFR* gene (specifically, an exon 19 deletion) and a *KRAS* G12C mutation. The question asks about the most appropriate next step in management. First, it’s crucial to recognize the presence of actionable driver mutations in NSCLC. *EGFR* mutations, such as exon 19 deletions or L858R point mutations, are well-established targets for tyrosine kinase inhibitors (TKIs). Similarly, *KRAS* mutations, particularly the G12C variant, have become targetable with the development of specific inhibitors. In a patient who has progressed on immunotherapy and chemotherapy, re-biopsy and molecular profiling are often indicated to identify new or persistent actionable mutations that can guide subsequent therapy. Given the documented *EGFR* exon 19 deletion, treatment with an *EGFR* TKI is the standard of care for such patients, as these agents have demonstrated superior efficacy and tolerability compared to chemotherapy in this setting. Osimertinib, a third-generation *EGFR* TKI, is particularly effective against both sensitizing *EGFR* mutations and the T790M resistance mutation, and it also has activity against *KRAS* mutations. While the patient also has a *KRAS* G12C mutation, which is also targetable, the *EGFR* mutation is generally considered the primary driver in this context, and *EGFR* TKIs are typically prioritized when both are present, especially after progression on immunotherapy. The development of resistance to prior treatments, including immunotherapy, necessitates a re-evaluation of the tumor’s molecular landscape. Therefore, initiating therapy with an *EGFR* TKI like osimertinib is the most evidence-based and effective approach to re-challenge the tumor and potentially achieve disease control. Other options, such as continuing chemotherapy or immunotherapy, are less likely to be effective given the documented progression on these modalities. Investigating other less common mutations without addressing the known driver mutations would be a suboptimal strategy.
-
Question 14 of 30
14. Question
A 68-year-old male, a former smoker with a 30-pack-year history, presents with progressive dyspnea and a persistent cough. Imaging reveals a large mass in the right upper lobe with mediastinal lymphadenopathy and distant hepatic metastases. A biopsy confirms poorly differentiated non-small cell lung cancer (NSCLC). Molecular profiling reveals a KRAS G12C mutation and no actionable mutations in EGFR, ALK, or ROS1. The patient has received two cycles of cisplatin and pembrolizumab, after which his disease has progressed based on RECIST criteria. Considering the molecular profile and treatment history, what is the most rational next therapeutic intervention for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C. This mutation is a common driver mutation in NSCLC and is targetable with specific small molecule inhibitors. The question asks about the most appropriate next therapeutic step. Given the documented efficacy and approval of KRAS G12C inhibitors in this setting, this represents the standard of care. The mechanism of action of these inhibitors involves blocking the constitutively active KRAS G12C protein, thereby inhibiting downstream signaling pathways that promote tumor growth and survival, such as the MAPK pathway. Other options are less appropriate. While continued chemotherapy might be considered in some scenarios, it is generally less effective after progression on prior lines, especially when a targetable mutation is present. Re-challenging with immunotherapy alone is unlikely to be effective given prior progression. Targeting EGFR mutations is irrelevant as the patient’s tumor does not harbor such mutations. Therefore, initiating therapy with a KRAS G12C inhibitor is the most evidence-based and effective approach for this patient.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C. This mutation is a common driver mutation in NSCLC and is targetable with specific small molecule inhibitors. The question asks about the most appropriate next therapeutic step. Given the documented efficacy and approval of KRAS G12C inhibitors in this setting, this represents the standard of care. The mechanism of action of these inhibitors involves blocking the constitutively active KRAS G12C protein, thereby inhibiting downstream signaling pathways that promote tumor growth and survival, such as the MAPK pathway. Other options are less appropriate. While continued chemotherapy might be considered in some scenarios, it is generally less effective after progression on prior lines, especially when a targetable mutation is present. Re-challenging with immunotherapy alone is unlikely to be effective given prior progression. Targeting EGFR mutations is irrelevant as the patient’s tumor does not harbor such mutations. Therefore, initiating therapy with a KRAS G12C inhibitor is the most evidence-based and effective approach for this patient.
-
Question 15 of 30
15. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Molecular profiling of his tumor reveals an activating *KRAS* G12C mutation and a high tumor mutational burden (TMB-H). He initially received a platinum-doublet chemotherapy regimen in combination with an immune checkpoint inhibitor, achieving a partial response. However, after 8 months of treatment, he developed progressive disease. Considering his clinical history and molecular findings, what is the most appropriate next therapeutic intervention for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the *KRAS* gene, specifically a G12C substitution, and also exhibits a high tumor mutational burden (TMB-H). The question asks for the most appropriate next therapeutic step. The patient has received prior platinum-based chemotherapy and an immune checkpoint inhibitor, and has now progressed. This indicates refractory disease to these standard treatments. The presence of a *KRAS* G12C mutation is a key actionable target. Sotorasib and adagrasib are approved targeted therapies specifically for NSCLC with *KRAS* G12C mutations. These agents inhibit the mutated KRAS protein, thereby blocking downstream signaling pathways that drive tumor growth. While TMB-H generally predicts a better response to immunotherapy, the patient has already progressed on immunotherapy, suggesting that further immune-based monotherapy might not be effective. Combination strategies involving immunotherapy and targeted therapy are being investigated, but for a patient with a confirmed *KRAS* G12C mutation, direct targeting of this driver mutation with a specific inhibitor is the most evidence-based and rational next step. Chemotherapy is a reasonable option for NSCLC, but given the presence of a targetable mutation, a targeted agent is preferred as it offers a more specific mechanism of action with potentially different toxicity profiles. Docetaxel is a microtubule inhibitor used in NSCLC, but it does not specifically address the underlying genetic driver in this case. Continuation of immunotherapy alone would be unlikely to yield significant benefit given the documented progression. Therefore, initiating therapy with a KRAS G12C inhibitor is the most appropriate management strategy.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the *KRAS* gene, specifically a G12C substitution, and also exhibits a high tumor mutational burden (TMB-H). The question asks for the most appropriate next therapeutic step. The patient has received prior platinum-based chemotherapy and an immune checkpoint inhibitor, and has now progressed. This indicates refractory disease to these standard treatments. The presence of a *KRAS* G12C mutation is a key actionable target. Sotorasib and adagrasib are approved targeted therapies specifically for NSCLC with *KRAS* G12C mutations. These agents inhibit the mutated KRAS protein, thereby blocking downstream signaling pathways that drive tumor growth. While TMB-H generally predicts a better response to immunotherapy, the patient has already progressed on immunotherapy, suggesting that further immune-based monotherapy might not be effective. Combination strategies involving immunotherapy and targeted therapy are being investigated, but for a patient with a confirmed *KRAS* G12C mutation, direct targeting of this driver mutation with a specific inhibitor is the most evidence-based and rational next step. Chemotherapy is a reasonable option for NSCLC, but given the presence of a targetable mutation, a targeted agent is preferred as it offers a more specific mechanism of action with potentially different toxicity profiles. Docetaxel is a microtubule inhibitor used in NSCLC, but it does not specifically address the underlying genetic driver in this case. Continuation of immunotherapy alone would be unlikely to yield significant benefit given the documented progression. Therefore, initiating therapy with a KRAS G12C inhibitor is the most appropriate management strategy.
-
Question 16 of 30
16. Question
A 68-year-old male presents with a new diagnosis of metastatic non-small cell lung cancer (NSCLC), adenocarcinoma histology. He received initial treatment with pembrolizumab and platinum-doublet chemotherapy, achieving a partial response. However, after 8 months of therapy, he developed progressive disease. Comprehensive genomic profiling of his tumor tissue revealed a KRAS G12C mutation, alongside a tumor mutational burden (TMB) of 15 mutations per megabase. Considering the patient’s clinical history and molecular findings, which of the following therapeutic interventions would represent the most targeted and potentially effective next step in management for this ABIM – Subspecialty in Medical Oncology University candidate to evaluate?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. The question asks about the most appropriate next step in management, considering the availability of targeted therapies. Sotorasib is a specific inhibitor of KRAS G12C, designed to block the downstream signaling pathways that promote tumor growth. While other options might be considered in different clinical contexts, sotorasib directly targets the identified molecular alteration, offering a rational approach to overcome resistance to prior therapies. Docetaxel is a standard chemotherapy agent, but without specific molecular rationale, it represents a less targeted approach. Continuation of immunotherapy is unlikely to be effective given documented progression on a prior immunotherapy regimen. Palliative radiation therapy might be considered for symptom control but does not address the underlying systemic disease progression driven by the KRAS mutation. Therefore, initiating treatment with a KRAS G12C inhibitor like sotorasib is the most evidence-based and targeted strategy in this specific clinical scenario, aligning with the principles of precision medicine emphasized in advanced oncology training at ABIM – Subspecialty in Medical Oncology University.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. The question asks about the most appropriate next step in management, considering the availability of targeted therapies. Sotorasib is a specific inhibitor of KRAS G12C, designed to block the downstream signaling pathways that promote tumor growth. While other options might be considered in different clinical contexts, sotorasib directly targets the identified molecular alteration, offering a rational approach to overcome resistance to prior therapies. Docetaxel is a standard chemotherapy agent, but without specific molecular rationale, it represents a less targeted approach. Continuation of immunotherapy is unlikely to be effective given documented progression on a prior immunotherapy regimen. Palliative radiation therapy might be considered for symptom control but does not address the underlying systemic disease progression driven by the KRAS mutation. Therefore, initiating treatment with a KRAS G12C inhibitor like sotorasib is the most evidence-based and targeted strategy in this specific clinical scenario, aligning with the principles of precision medicine emphasized in advanced oncology training at ABIM – Subspecialty in Medical Oncology University.
-
Question 17 of 30
17. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC) with adenocarcinoma histology. Initial treatment with pembrolizumab and carboplatin/pemetrexed resulted in a partial response, but after 8 months, he developed progressive disease. Comprehensive genomic profiling of his tumor tissue revealed a KRAS G12C mutation. Considering the principles of personalized therapy and the advancements in molecularly targeted agents, which of the following therapeutic strategies would be most appropriate for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C. The question asks for the most appropriate next therapeutic step. Given the documented efficacy and regulatory approval of KRAS G12C inhibitors in this setting, this represents the most targeted and potentially beneficial approach. The rationale for selecting a KRAS G12C inhibitor is based on its direct mechanism of action, which is to inhibit the constitutively active signaling pathway driven by the mutation. This is a prime example of precision medicine in oncology, a core principle emphasized at ABIM – Subspecialty in Medical Oncology University. Other options are less optimal: continuing non-specific chemotherapy might offer limited benefit after progression on a similar regimen; switching to a different immunotherapy without a clear predictive biomarker for response in this context is less likely to be effective; and a broad panel next-generation sequencing (NGS) test, while valuable for identifying other potential targets, does not directly dictate the immediate next step when a specific actionable mutation like KRAS G12C is already known. The explanation emphasizes the importance of identifying and targeting specific molecular alterations in cancer treatment, a cornerstone of modern medical oncology.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C. The question asks for the most appropriate next therapeutic step. Given the documented efficacy and regulatory approval of KRAS G12C inhibitors in this setting, this represents the most targeted and potentially beneficial approach. The rationale for selecting a KRAS G12C inhibitor is based on its direct mechanism of action, which is to inhibit the constitutively active signaling pathway driven by the mutation. This is a prime example of precision medicine in oncology, a core principle emphasized at ABIM – Subspecialty in Medical Oncology University. Other options are less optimal: continuing non-specific chemotherapy might offer limited benefit after progression on a similar regimen; switching to a different immunotherapy without a clear predictive biomarker for response in this context is less likely to be effective; and a broad panel next-generation sequencing (NGS) test, while valuable for identifying other potential targets, does not directly dictate the immediate next step when a specific actionable mutation like KRAS G12C is already known. The explanation emphasizes the importance of identifying and targeting specific molecular alterations in cancer treatment, a cornerstone of modern medical oncology.
-
Question 18 of 30
18. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC), adenocarcinoma histology. He received first-line pembrolizumab and carboplatin, with initial partial response followed by progressive disease after 8 months. Comprehensive genomic profiling of his tumor tissue reveals a KRAS G12C mutation, PD-L1 TPS of 5%, and no EGFR, ALK, or ROS1 alterations. Considering the molecular findings and treatment history, which of the following represents the most appropriate next-line systemic therapy for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C. The question probes the appropriate next-line therapy considering the molecular profile of the tumor and the patient’s treatment history. Current guidelines and clinical trial data support the use of KRAS G12C inhibitors as a targeted therapy option for patients with this specific mutation after progression on standard first-line treatments. Sotorasib and adagrasib are examples of such inhibitors. Docetaxel is a standard chemotherapy agent, but given the presence of a targetable mutation and prior immunotherapy, a targeted approach is generally preferred. Continuation of immunotherapy alone is unlikely to be effective given prior progression. Palliative radiation therapy might be considered for specific symptomatic sites but is not a systemic treatment for the overall disease burden. Therefore, initiating a KRAS G12C inhibitor represents the most evidence-based and targeted approach to manage this patient’s disease.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene, specifically KRAS G12C. The question probes the appropriate next-line therapy considering the molecular profile of the tumor and the patient’s treatment history. Current guidelines and clinical trial data support the use of KRAS G12C inhibitors as a targeted therapy option for patients with this specific mutation after progression on standard first-line treatments. Sotorasib and adagrasib are examples of such inhibitors. Docetaxel is a standard chemotherapy agent, but given the presence of a targetable mutation and prior immunotherapy, a targeted approach is generally preferred. Continuation of immunotherapy alone is unlikely to be effective given prior progression. Palliative radiation therapy might be considered for specific symptomatic sites but is not a systemic treatment for the overall disease burden. Therefore, initiating a KRAS G12C inhibitor represents the most evidence-based and targeted approach to manage this patient’s disease.
-
Question 19 of 30
19. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Initial treatment with pembrolizumab and carboplatin resulted in a partial response for 10 months, followed by progressive disease. Comprehensive genomic profiling of the tumor tissue revealed a KRAS G12C mutation, along with a PD-L1 tumor proportion score of 60%. Considering the patient’s clinical history and molecular findings, which of the following therapeutic strategies would be most appropriate as the next line of treatment at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The question probes the understanding of resistance mechanisms and appropriate subsequent treatment strategies, a core competency for ABIM – Subspecialty in Medical Oncology University candidates. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. While the initial immunotherapy regimen (likely an immune checkpoint inhibitor) may have been effective, resistance can develop through various mechanisms, including alterations in antigen presentation, T-cell exhaustion, or the tumor microenvironment. The KRAS G12C mutation itself can contribute to tumor growth and survival, and its presence suggests a potential target for specific therapies. The development of a KRAS G12C inhibitor represents a significant advancement in personalized oncology for NSCLC. These inhibitors are designed to specifically block the aberrant signaling pathway driven by the mutated KRAS protein, thereby inhibiting tumor cell proliferation and survival. The clinical efficacy of these agents has been demonstrated in patients with KRAS G12C-mutated NSCLC who have progressed on prior standard therapies. Therefore, introducing a KRAS G12C inhibitor is a logical and evidence-based next step in this patient’s treatment. Other options are less appropriate. Continued immunotherapy without addressing the identified KRAS mutation might not overcome resistance mechanisms. Chemotherapy, while an option, may offer less targeted efficacy and a different toxicity profile compared to a specific molecular inhibitor. Investigating other driver mutations is important, but the presence of a confirmed KRAS G12C mutation warrants targeting it directly.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The question probes the understanding of resistance mechanisms and appropriate subsequent treatment strategies, a core competency for ABIM – Subspecialty in Medical Oncology University candidates. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. While the initial immunotherapy regimen (likely an immune checkpoint inhibitor) may have been effective, resistance can develop through various mechanisms, including alterations in antigen presentation, T-cell exhaustion, or the tumor microenvironment. The KRAS G12C mutation itself can contribute to tumor growth and survival, and its presence suggests a potential target for specific therapies. The development of a KRAS G12C inhibitor represents a significant advancement in personalized oncology for NSCLC. These inhibitors are designed to specifically block the aberrant signaling pathway driven by the mutated KRAS protein, thereby inhibiting tumor cell proliferation and survival. The clinical efficacy of these agents has been demonstrated in patients with KRAS G12C-mutated NSCLC who have progressed on prior standard therapies. Therefore, introducing a KRAS G12C inhibitor is a logical and evidence-based next step in this patient’s treatment. Other options are less appropriate. Continued immunotherapy without addressing the identified KRAS mutation might not overcome resistance mechanisms. Chemotherapy, while an option, may offer less targeted efficacy and a different toxicity profile compared to a specific molecular inhibitor. Investigating other driver mutations is important, but the presence of a confirmed KRAS G12C mutation warrants targeting it directly.
-
Question 20 of 30
20. Question
A 68-year-old male with a history of smoking presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Initial treatment with carboplatin, pemetrexed, and pembrolizumab resulted in a partial response, but after 8 months, progressive disease was confirmed on imaging. Comprehensive genomic profiling of the tumor tissue revealed a KRAS G12C mutation. The patient is performance status 1 and has no significant comorbidities. Considering the molecular findings and prior treatment failure, what is the most appropriate next therapeutic intervention for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, and the physician is considering a targeted therapy. The question asks about the most appropriate next step in management, considering the molecular profile and prior treatment. The patient has received prior platinum-based chemotherapy and an immune checkpoint inhibitor, and has now progressed. The tumor is confirmed to have a KRAS G12C mutation. Sotorasib is a specific inhibitor of KRAS G12C. Clinical trials and subsequent approvals have demonstrated efficacy of KRAS G12C inhibitors in patients with this specific mutation who have progressed on prior therapy. Therefore, initiating treatment with sotorasib is the most evidence-based and targeted approach for this patient. Other options are less appropriate. Continuing or switching to another non-specific chemotherapy regimen without addressing the actionable KRAS G12C mutation would be suboptimal. Re-challenging with immunotherapy alone, without considering the molecular context or potential resistance mechanisms, is unlikely to be as effective as targeted therapy. While a biopsy for further molecular profiling is always an option, it is not the immediate next step when a known actionable mutation like KRAS G12C is present and a targeted therapy is available. The primary goal is to leverage the identified molecular alteration for a more effective treatment.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, and the physician is considering a targeted therapy. The question asks about the most appropriate next step in management, considering the molecular profile and prior treatment. The patient has received prior platinum-based chemotherapy and an immune checkpoint inhibitor, and has now progressed. The tumor is confirmed to have a KRAS G12C mutation. Sotorasib is a specific inhibitor of KRAS G12C. Clinical trials and subsequent approvals have demonstrated efficacy of KRAS G12C inhibitors in patients with this specific mutation who have progressed on prior therapy. Therefore, initiating treatment with sotorasib is the most evidence-based and targeted approach for this patient. Other options are less appropriate. Continuing or switching to another non-specific chemotherapy regimen without addressing the actionable KRAS G12C mutation would be suboptimal. Re-challenging with immunotherapy alone, without considering the molecular context or potential resistance mechanisms, is unlikely to be as effective as targeted therapy. While a biopsy for further molecular profiling is always an option, it is not the immediate next step when a known actionable mutation like KRAS G12C is present and a targeted therapy is available. The primary goal is to leverage the identified molecular alteration for a more effective treatment.
-
Question 21 of 30
21. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Initial treatment with pembrolizumab and carboplatin/pemetrexed resulted in a partial response, but after 8 months, he developed progressive disease. Comprehensive genomic profiling of his tumor tissue revealed a KRAS G12C mutation and a high tumor mutational burden (TMB-H). Given this molecular profile and his treatment history, which of the following represents the most appropriate next therapeutic intervention for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, and molecular profiling also reveals a high tumor mutational burden (TMB-H). The question asks for the most appropriate next therapeutic step. Considering the patient’s progression after prior treatments and the identified molecular alterations, the optimal strategy involves leveraging targeted therapy for the KRAS G12C mutation, as this represents a direct actionable target. Sotorasib and adagrasib are approved KRAS G12C inhibitors that have demonstrated efficacy in this patient population. While TMB-H can predict response to immunotherapy, the patient has already progressed on immunotherapy, making further immune checkpoint inhibition alone less likely to be effective without a novel combination or different immune-modulating strategy. Continuation of chemotherapy would not address the specific driver mutation. Radiation therapy is typically reserved for localized disease or palliation of specific metastatic sites, not as a systemic treatment for widespread progression. Therefore, initiating a KRAS G12C inhibitor is the most targeted and evidence-based approach to address the underlying oncogenic driver in this context, aligning with the principles of precision medicine emphasized at ABIM – Subspecialty in Medical Oncology University.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, and molecular profiling also reveals a high tumor mutational burden (TMB-H). The question asks for the most appropriate next therapeutic step. Considering the patient’s progression after prior treatments and the identified molecular alterations, the optimal strategy involves leveraging targeted therapy for the KRAS G12C mutation, as this represents a direct actionable target. Sotorasib and adagrasib are approved KRAS G12C inhibitors that have demonstrated efficacy in this patient population. While TMB-H can predict response to immunotherapy, the patient has already progressed on immunotherapy, making further immune checkpoint inhibition alone less likely to be effective without a novel combination or different immune-modulating strategy. Continuation of chemotherapy would not address the specific driver mutation. Radiation therapy is typically reserved for localized disease or palliation of specific metastatic sites, not as a systemic treatment for widespread progression. Therefore, initiating a KRAS G12C inhibitor is the most targeted and evidence-based approach to address the underlying oncogenic driver in this context, aligning with the principles of precision medicine emphasized at ABIM – Subspecialty in Medical Oncology University.
-
Question 22 of 30
22. Question
A 68-year-old male with metastatic non-small cell lung cancer (NSCLC) has progressed following first-line platinum-based chemotherapy. Molecular profiling of his tumor biopsy reveals no actionable driver mutations. He is now being considered for treatment with an immune checkpoint inhibitor (ICI), specifically pembrolizumab. The histopathological analysis of the tumor biopsy demonstrates a TME characterized by a high infiltrate of CD8+ cytotoxic T lymphocytes, a notable population of FOXP3+ regulatory T cells, and a pronounced desmoplastic stromal reaction. Given this immunophenotype of the tumor microenvironment, what is the most likely clinical implication regarding his potential response to pembrolizumab?
Correct
The question probes the understanding of the interplay between tumor microenvironment (TME) components and the efficacy of immune checkpoint inhibitors (ICIs), a core concept in modern medical oncology relevant to ABIM – Subspecialty in Medical Oncology University’s curriculum. Specifically, it focuses on how the presence of specific immune cells and stromal elements within the TME can predict or influence response to ICIs. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on standard chemotherapy and is being considered for pembrolizumab. The biopsy reveals a TME characterized by a high density of tumor-infiltrating lymphocytes (TILs), particularly CD8+ T cells, a moderate presence of regulatory T cells (Tregs), and a dense desmoplastic stroma. To answer this question, one must recall the established prognostic and predictive roles of these TME components in ICI therapy. A high density of CD8+ TILs is generally associated with a favorable response to ICIs, as these cytotoxic T cells are the primary effectors of anti-tumor immunity. Tregs, while present in the TME, can suppress anti-tumor immune responses, and their high prevalence can sometimes be linked to resistance. A dense desmoplastic stroma can act as a physical barrier, hindering immune cell infiltration and function, and is often associated with poorer outcomes in ICI therapy. Therefore, the combination of abundant CD8+ TILs and a dense desmoplastic stroma presents a complex scenario. While the CD8+ TILs suggest a potential for response, the desmoplastic stroma poses a significant challenge to immune cell penetration and activity. The presence of Tregs further complicates the picture by potentially dampening the immune response. Considering these factors, the most accurate assessment of the patient’s likely response to pembrolizumab, based on the described TME, would be a guarded prognosis, with a possibility of response but significant hurdles to overcome. This nuanced understanding of TME composition and its impact on immunotherapy efficacy is crucial for advanced oncology trainees at ABIM – Subspecialty in Medical Oncology University.
Incorrect
The question probes the understanding of the interplay between tumor microenvironment (TME) components and the efficacy of immune checkpoint inhibitors (ICIs), a core concept in modern medical oncology relevant to ABIM – Subspecialty in Medical Oncology University’s curriculum. Specifically, it focuses on how the presence of specific immune cells and stromal elements within the TME can predict or influence response to ICIs. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) who has progressed on standard chemotherapy and is being considered for pembrolizumab. The biopsy reveals a TME characterized by a high density of tumor-infiltrating lymphocytes (TILs), particularly CD8+ T cells, a moderate presence of regulatory T cells (Tregs), and a dense desmoplastic stroma. To answer this question, one must recall the established prognostic and predictive roles of these TME components in ICI therapy. A high density of CD8+ TILs is generally associated with a favorable response to ICIs, as these cytotoxic T cells are the primary effectors of anti-tumor immunity. Tregs, while present in the TME, can suppress anti-tumor immune responses, and their high prevalence can sometimes be linked to resistance. A dense desmoplastic stroma can act as a physical barrier, hindering immune cell infiltration and function, and is often associated with poorer outcomes in ICI therapy. Therefore, the combination of abundant CD8+ TILs and a dense desmoplastic stroma presents a complex scenario. While the CD8+ TILs suggest a potential for response, the desmoplastic stroma poses a significant challenge to immune cell penetration and activity. The presence of Tregs further complicates the picture by potentially dampening the immune response. Considering these factors, the most accurate assessment of the patient’s likely response to pembrolizumab, based on the described TME, would be a guarded prognosis, with a possibility of response but significant hurdles to overcome. This nuanced understanding of TME composition and its impact on immunotherapy efficacy is crucial for advanced oncology trainees at ABIM – Subspecialty in Medical Oncology University.
-
Question 23 of 30
23. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Molecular profiling of his tumor reveals an activating *KRAS* G12C mutation and a high tumor mutational burden (TMB-H). He initially received a platinum-based chemotherapy regimen combined with an immune checkpoint inhibitor, achieving a partial response. However, after 8 months of treatment, he experienced disease progression. Considering the patient’s clinical and molecular profile, and the established advancements in personalized oncology taught at ABIM – Subspecialty in Medical Oncology University, what is the most appropriate next therapeutic intervention?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the *KRAS* gene, specifically a G12C substitution, and also exhibits a high tumor mutational burden (TMB-H). The question asks for the most appropriate next therapeutic step. First, let’s analyze the patient’s profile. The presence of a *KRAS* G12C mutation in NSCLC is a significant actionable target. Sotorasib and adagrasib are approved targeted therapies specifically designed to inhibit this mutated protein. Given the progression on prior lines of therapy, including immunotherapy, a targeted approach is warranted. Second, the high TMB (TMB-H) is generally associated with a better response to immune checkpoint inhibitors. However, the patient has already progressed on immunotherapy, suggesting that either the initial response was transient or the tumor has developed resistance mechanisms. While TMB-H can predict response, it doesn’t negate the efficacy of targeted therapy when an actionable mutation is present. Considering the available treatment options for *KRAS* G12C-mutated NSCLC after progression on chemotherapy and immunotherapy, a *KRAS* G12C inhibitor like sotorasib or adagrasib is the most evidence-based and effective next step. These agents directly inhibit the constitutively active mutated KRAS protein, leading to downstream pathway inhibition and tumor growth suppression. Therefore, the correct approach is to initiate treatment with a *KRAS* G12C inhibitor. This strategy leverages the specific molecular alteration in the tumor to provide a more personalized and potentially effective treatment, aligning with the principles of precision medicine emphasized in advanced oncology training at ABIM – Subspecialty in Medical Oncology University. The other options are less optimal. Continuing or re-challenging with immunotherapy might be considered in specific contexts, but not as the immediate next step given prior progression. Chemotherapy, while an option, is generally less effective than targeted therapy in this specific molecular subset after initial failure. Investigating other driver mutations is important for future treatment planning but does not represent the immediate best next step for a *KRAS* G12C-mutated tumor.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the *KRAS* gene, specifically a G12C substitution, and also exhibits a high tumor mutational burden (TMB-H). The question asks for the most appropriate next therapeutic step. First, let’s analyze the patient’s profile. The presence of a *KRAS* G12C mutation in NSCLC is a significant actionable target. Sotorasib and adagrasib are approved targeted therapies specifically designed to inhibit this mutated protein. Given the progression on prior lines of therapy, including immunotherapy, a targeted approach is warranted. Second, the high TMB (TMB-H) is generally associated with a better response to immune checkpoint inhibitors. However, the patient has already progressed on immunotherapy, suggesting that either the initial response was transient or the tumor has developed resistance mechanisms. While TMB-H can predict response, it doesn’t negate the efficacy of targeted therapy when an actionable mutation is present. Considering the available treatment options for *KRAS* G12C-mutated NSCLC after progression on chemotherapy and immunotherapy, a *KRAS* G12C inhibitor like sotorasib or adagrasib is the most evidence-based and effective next step. These agents directly inhibit the constitutively active mutated KRAS protein, leading to downstream pathway inhibition and tumor growth suppression. Therefore, the correct approach is to initiate treatment with a *KRAS* G12C inhibitor. This strategy leverages the specific molecular alteration in the tumor to provide a more personalized and potentially effective treatment, aligning with the principles of precision medicine emphasized in advanced oncology training at ABIM – Subspecialty in Medical Oncology University. The other options are less optimal. Continuing or re-challenging with immunotherapy might be considered in specific contexts, but not as the immediate next step given prior progression. Chemotherapy, while an option, is generally less effective than targeted therapy in this specific molecular subset after initial failure. Investigating other driver mutations is important for future treatment planning but does not represent the immediate best next step for a *KRAS* G12C-mutated tumor.
-
Question 24 of 30
24. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). He undergoes initial treatment with carboplatin and pembrolizumab, achieving a partial response. However, after 8 months, he develops progressive disease. Comprehensive genomic profiling of his tumor reveals a KRAS G12C mutation, along with a tumor mutational burden (TMB) of 15 mutations per megabase. Considering his treatment history and molecular findings, what is the most appropriate next therapeutic intervention to consider for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a specific genetic alteration: a KRAS G12C mutation. The question asks about the most appropriate next therapeutic step, considering the molecular profile and prior treatment. The patient has received a platinum-based doublet and an immune checkpoint inhibitor, and has now progressed. This indicates resistance to standard first-line therapy. The presence of a KRAS G12C mutation is a key actionable target. Recent advancements in targeted therapy have led to the development of specific inhibitors for this mutation. Sotorasib and adagrasib are examples of such inhibitors that have demonstrated clinical activity in patients with KRAS G12C-mutated NSCLC who have progressed after prior therapy. Therefore, initiating treatment with a KRAS G12C inhibitor is the most rational next step. Other options are less appropriate. Continuing chemotherapy alone without a targeted agent may offer limited benefit given the progression. Re-challenging with immunotherapy might be considered in specific contexts, but the patient has already progressed on immunotherapy, and a targeted agent is available for a known driver mutation. Switching to a different class of chemotherapy without a targeted approach is also less optimal than leveraging the identified molecular vulnerability.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a specific genetic alteration: a KRAS G12C mutation. The question asks about the most appropriate next therapeutic step, considering the molecular profile and prior treatment. The patient has received a platinum-based doublet and an immune checkpoint inhibitor, and has now progressed. This indicates resistance to standard first-line therapy. The presence of a KRAS G12C mutation is a key actionable target. Recent advancements in targeted therapy have led to the development of specific inhibitors for this mutation. Sotorasib and adagrasib are examples of such inhibitors that have demonstrated clinical activity in patients with KRAS G12C-mutated NSCLC who have progressed after prior therapy. Therefore, initiating treatment with a KRAS G12C inhibitor is the most rational next step. Other options are less appropriate. Continuing chemotherapy alone without a targeted agent may offer limited benefit given the progression. Re-challenging with immunotherapy might be considered in specific contexts, but the patient has already progressed on immunotherapy, and a targeted agent is available for a known driver mutation. Switching to a different class of chemotherapy without a targeted approach is also less optimal than leveraging the identified molecular vulnerability.
-
Question 25 of 30
25. Question
A 68-year-old male diagnosed with stage IV non-small cell lung cancer (NSCLC) is initiated on pembrolizumab monotherapy. Prior to treatment, a tumor biopsy was performed. Histopathological and immunohistochemical analysis of the biopsy specimen reveals a tumor microenvironment characterized by a significant infiltrate of CD4+ FOXP3+ regulatory T cells (Tregs) within the tumor stroma, a high density of alpha-smooth muscle actin (α-SMA) positive cancer-associated fibroblasts (CAFs), and a marked scarcity of CD8+ cytotoxic T lymphocytes (CTLs) within the tumor nests. Based on these findings and the known mechanisms of immune evasion, which of the following is the most likely clinical outcome regarding response to pembrolizumab?
Correct
The question probes the understanding of the interplay between tumor microenvironment (TME) components and the efficacy of immune checkpoint inhibitors (ICIs). Specifically, it focuses on how the presence of specific immune cells and stromal elements within the TME can predict response to ICIs, a critical concept in modern oncology practice relevant to ABIM – Subspecialty in Medical Oncology University’s curriculum. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) treated with an anti-PD-1 antibody. The tumor biopsy reveals a dense stromal infiltrate of regulatory T cells (Tregs), abundant immunosuppressive fibroblasts (CAFs), and a paucity of cytotoxic T lymphocytes (CTLs) within the tumor nests. This cellular and molecular profile is indicative of an immune-excluded or immune-desert phenotype, characterized by a hostile TME that actively suppresses anti-tumor immunity. Tregs are known to dampen the activity of effector T cells, including CTLs, through various mechanisms such as secreting immunosuppressive cytokines (e.g., IL-10, TGF-β) and direct cell-to-cell contact. CAFs contribute to immune suppression by secreting immunosuppressive factors, remodeling the extracellular matrix to impede immune cell infiltration, and promoting angiogenesis that can further isolate tumor cells. A lack of CTLs within the tumor parenchyma signifies a failure of these effector cells to infiltrate and recognize tumor antigens, a prerequisite for effective ICI therapy. Therefore, a TME rich in Tregs and CAFs, with a deficit of intratumoral CTLs, is strongly associated with primary resistance to ICIs. This is because the fundamental mechanism of ICIs is to unleash pre-existing or newly activated anti-tumor T cell responses by blocking inhibitory signals (like PD-1/PD-L1). If the TME is fundamentally immunosuppressive and lacks effector T cells capable of recognizing the tumor, ICIs will have limited impact. The correct approach to managing such a patient would involve considering alternative or combination strategies that can overcome this immunosuppressive TME. This might include agents that deplete Tregs, target CAFs, or enhance T cell infiltration and activation, potentially in combination with ICIs. Understanding these TME characteristics is paramount for personalized treatment selection and is a core competency emphasized at ABIM – Subspecialty in Medical Oncology University.
Incorrect
The question probes the understanding of the interplay between tumor microenvironment (TME) components and the efficacy of immune checkpoint inhibitors (ICIs). Specifically, it focuses on how the presence of specific immune cells and stromal elements within the TME can predict response to ICIs, a critical concept in modern oncology practice relevant to ABIM – Subspecialty in Medical Oncology University’s curriculum. The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) treated with an anti-PD-1 antibody. The tumor biopsy reveals a dense stromal infiltrate of regulatory T cells (Tregs), abundant immunosuppressive fibroblasts (CAFs), and a paucity of cytotoxic T lymphocytes (CTLs) within the tumor nests. This cellular and molecular profile is indicative of an immune-excluded or immune-desert phenotype, characterized by a hostile TME that actively suppresses anti-tumor immunity. Tregs are known to dampen the activity of effector T cells, including CTLs, through various mechanisms such as secreting immunosuppressive cytokines (e.g., IL-10, TGF-β) and direct cell-to-cell contact. CAFs contribute to immune suppression by secreting immunosuppressive factors, remodeling the extracellular matrix to impede immune cell infiltration, and promoting angiogenesis that can further isolate tumor cells. A lack of CTLs within the tumor parenchyma signifies a failure of these effector cells to infiltrate and recognize tumor antigens, a prerequisite for effective ICI therapy. Therefore, a TME rich in Tregs and CAFs, with a deficit of intratumoral CTLs, is strongly associated with primary resistance to ICIs. This is because the fundamental mechanism of ICIs is to unleash pre-existing or newly activated anti-tumor T cell responses by blocking inhibitory signals (like PD-1/PD-L1). If the TME is fundamentally immunosuppressive and lacks effector T cells capable of recognizing the tumor, ICIs will have limited impact. The correct approach to managing such a patient would involve considering alternative or combination strategies that can overcome this immunosuppressive TME. This might include agents that deplete Tregs, target CAFs, or enhance T cell infiltration and activation, potentially in combination with ICIs. Understanding these TME characteristics is paramount for personalized treatment selection and is a core competency emphasized at ABIM – Subspecialty in Medical Oncology University.
-
Question 26 of 30
26. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Molecular profiling of his tumor reveals a \(KRAS^{G12C}\) mutation, amplification of \(EGFR\), and a PD-L1 tumor proportion score of 60%. He initially receives a combination of pembrolizumab and platinum-doublet chemotherapy, achieving a partial response. However, after 8 months, he experiences documented disease progression. Considering the molecular profile and prior treatment history, which of the following represents the most rational next therapeutic intervention for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the \(KRAS\) gene, specifically \(KRAS^{G12C}\). This mutation leads to a constitutively active RAS protein, which drives downstream signaling pathways such as the MAPK pathway, promoting cell proliferation and survival. The question asks for the most appropriate next therapeutic step. Given the presence of a \(KRAS^{G12C}\) mutation and progression on prior therapies, a targeted therapy directed against this specific mutation is the most logical and evidence-based approach. Sotorasib and adagrasib are approved covalent inhibitors of \(KRAS^{G12C}\) that bind to the mutated protein and lock it in an inactive conformation, thereby inhibiting downstream signaling. Other options are less suitable: – Continuation of platinum-based chemotherapy would likely be ineffective given prior progression and the availability of a targeted agent. – Re-challenge with immunotherapy might be considered in some scenarios, but the presence of a targetable mutation makes targeted therapy the preferred next step, especially after progression on immunotherapy. – Radiation therapy to a specific metastatic site might be used for palliation of symptoms but is not a systemic treatment for widespread metastatic disease and does not address the underlying molecular driver. Therefore, initiating treatment with a \(KRAS^{G12C}\) inhibitor is the most appropriate management strategy in this context, aligning with precision medicine principles emphasized at ABIM – Subspecialty in Medical Oncology University.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the \(KRAS\) gene, specifically \(KRAS^{G12C}\). This mutation leads to a constitutively active RAS protein, which drives downstream signaling pathways such as the MAPK pathway, promoting cell proliferation and survival. The question asks for the most appropriate next therapeutic step. Given the presence of a \(KRAS^{G12C}\) mutation and progression on prior therapies, a targeted therapy directed against this specific mutation is the most logical and evidence-based approach. Sotorasib and adagrasib are approved covalent inhibitors of \(KRAS^{G12C}\) that bind to the mutated protein and lock it in an inactive conformation, thereby inhibiting downstream signaling. Other options are less suitable: – Continuation of platinum-based chemotherapy would likely be ineffective given prior progression and the availability of a targeted agent. – Re-challenge with immunotherapy might be considered in some scenarios, but the presence of a targetable mutation makes targeted therapy the preferred next step, especially after progression on immunotherapy. – Radiation therapy to a specific metastatic site might be used for palliation of symptoms but is not a systemic treatment for widespread metastatic disease and does not address the underlying molecular driver. Therefore, initiating treatment with a \(KRAS^{G12C}\) inhibitor is the most appropriate management strategy in this context, aligning with precision medicine principles emphasized at ABIM – Subspecialty in Medical Oncology University.
-
Question 27 of 30
27. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Molecular profiling of his tumor reveals a KRAS G12C mutation and a programmed death-ligand 1 (PD-L1) expression of 60%. He initially received a platinum-based chemotherapy regimen combined with pembrolizumab, but experienced disease progression after six cycles. Considering the patient’s clinical presentation and molecular findings, which of the following therapeutic strategies would be most aligned with current precision oncology principles for ABIM – Subspecialty in Medical Oncology University’s advanced curriculum?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene (specifically, KRAS G12C) and also expresses PD-L1 at 60%. The question asks about the most appropriate next therapeutic step. Given the documented KRAS G12C mutation, a targeted therapy specifically designed to inhibit this mutated protein is a highly effective option. Sotorasib is a covalent inhibitor of KRAS G12C that has demonstrated clinical activity in patients with this specific mutation. While continued immunotherapy might be considered in some contexts, the progression on prior immunotherapy suggests a diminished likelihood of significant benefit from further PD-1/PD-L1 blockade alone, especially when a targetable driver mutation is present. Chemotherapy, while an option, is generally considered less effective than targeted therapy when a specific actionable mutation is identified. Palliative radiation therapy is indicated for symptom management, not as a systemic treatment for widespread metastatic disease in this context. Therefore, initiating treatment with a KRAS G12C inhibitor represents the most precise and potentially beneficial therapeutic strategy based on the molecular profile of the tumor.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on initial platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene (specifically, KRAS G12C) and also expresses PD-L1 at 60%. The question asks about the most appropriate next therapeutic step. Given the documented KRAS G12C mutation, a targeted therapy specifically designed to inhibit this mutated protein is a highly effective option. Sotorasib is a covalent inhibitor of KRAS G12C that has demonstrated clinical activity in patients with this specific mutation. While continued immunotherapy might be considered in some contexts, the progression on prior immunotherapy suggests a diminished likelihood of significant benefit from further PD-1/PD-L1 blockade alone, especially when a targetable driver mutation is present. Chemotherapy, while an option, is generally considered less effective than targeted therapy when a specific actionable mutation is identified. Palliative radiation therapy is indicated for symptom management, not as a systemic treatment for widespread metastatic disease in this context. Therefore, initiating treatment with a KRAS G12C inhibitor represents the most precise and potentially beneficial therapeutic strategy based on the molecular profile of the tumor.
-
Question 28 of 30
28. Question
A 68-year-old male presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC) with pleural effusions and bone metastases. Initial treatment with carboplatin, pemetrexed, and pembrolizumab resulted in a partial response for 6 months, followed by progressive disease in the pleura and development of new bone lesions. Comprehensive genomic profiling of the tumor tissue reveals a KRAS G12C mutation, a PD-L1 tumor proportion score of 70%, and no other actionable mutations. Considering the patient’s disease progression and the identified molecular alteration, what is the most appropriate subsequent therapeutic intervention for this patient at ABIM – Subspecialty in Medical Oncology University?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. The question asks about the most appropriate next step in management. Sotorasib is a targeted therapy specifically designed to inhibit the KRAS G12C protein, which is constitutively active in cancer cells, driving uncontrolled proliferation. Therefore, administering sotorasib is the most logical and evidence-based approach for a patient with a confirmed KRAS G12C mutation who has progressed on standard therapies. Other options are less appropriate. Continuing platinum-based chemotherapy after progression is generally not recommended as it is unlikely to be effective and may increase toxicity. Docetaxel is a standard chemotherapy agent, but without specific molecular targets identified, its efficacy might be limited compared to a targeted agent. Palliative radiation therapy to the bone metastasis is appropriate for symptom management but does not address the underlying systemic disease progression driven by the KRAS mutation. While a comprehensive genomic profiling might be considered in some contexts, in this specific scenario, the presence of a known actionable mutation (KRAS G12C) directs the immediate therapeutic decision. The ABIM – Subspecialty in Medical Oncology University emphasizes evidence-based practice and the application of molecular diagnostics to guide treatment, making the targeted approach the most aligned with its educational principles.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors a KRAS G12C mutation, a common oncogenic driver in NSCLC. The question asks about the most appropriate next step in management. Sotorasib is a targeted therapy specifically designed to inhibit the KRAS G12C protein, which is constitutively active in cancer cells, driving uncontrolled proliferation. Therefore, administering sotorasib is the most logical and evidence-based approach for a patient with a confirmed KRAS G12C mutation who has progressed on standard therapies. Other options are less appropriate. Continuing platinum-based chemotherapy after progression is generally not recommended as it is unlikely to be effective and may increase toxicity. Docetaxel is a standard chemotherapy agent, but without specific molecular targets identified, its efficacy might be limited compared to a targeted agent. Palliative radiation therapy to the bone metastasis is appropriate for symptom management but does not address the underlying systemic disease progression driven by the KRAS mutation. While a comprehensive genomic profiling might be considered in some contexts, in this specific scenario, the presence of a known actionable mutation (KRAS G12C) directs the immediate therapeutic decision. The ABIM – Subspecialty in Medical Oncology University emphasizes evidence-based practice and the application of molecular diagnostics to guide treatment, making the targeted approach the most aligned with its educational principles.
-
Question 29 of 30
29. Question
A 68-year-old male presents with metastatic non-small cell lung cancer (NSCLC) that has progressed after first-line platinum-doublet chemotherapy and subsequent pembrolizumab therapy. Comprehensive genomic profiling of his tumor reveals an activating KRAS G12C mutation and a concurrent inactivating mutation in STK11. Given this molecular profile and the patient’s treatment history, what is the most likely clinical outcome if he is treated with a KRAS G12C-specific inhibitor?
Correct
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene (specifically, KRAS G12C) and a co-occurring mutation in STK11. KRAS G12C is a targetable mutation, and specific inhibitors like sotorasib and adagrasib are available. However, STK11 mutations are known to be associated with resistance to immunotherapy, particularly PD-1/PD-L1 blockade, and can also influence response to KRAS inhibitors. Studies have shown that patients with STK11 mutations may have a poorer response to KRAS G12C inhibitors compared to those without this co-mutation. Therefore, while a KRAS G12C inhibitor is a logical next step given the molecular profile, the presence of the STK11 mutation suggests a potentially diminished efficacy and a higher likelihood of resistance mechanisms emerging. Considering the options, a KRAS G12C inhibitor is the most appropriate targeted therapy. However, the question asks for the *most likely* outcome considering the molecular context. The combination of KRAS G12C and STK11 mutations often predicts a less robust response to KRAS inhibitors, and the patient has already progressed on immunotherapy, suggesting a limited benefit from further immune checkpoint blockade alone. Therefore, the most accurate prediction is a limited duration of response.
Incorrect
The scenario describes a patient with metastatic non-small cell lung cancer (NSCLC) who has progressed on platinum-based chemotherapy and immunotherapy. The patient’s tumor harbors an activating mutation in the KRAS gene (specifically, KRAS G12C) and a co-occurring mutation in STK11. KRAS G12C is a targetable mutation, and specific inhibitors like sotorasib and adagrasib are available. However, STK11 mutations are known to be associated with resistance to immunotherapy, particularly PD-1/PD-L1 blockade, and can also influence response to KRAS inhibitors. Studies have shown that patients with STK11 mutations may have a poorer response to KRAS G12C inhibitors compared to those without this co-mutation. Therefore, while a KRAS G12C inhibitor is a logical next step given the molecular profile, the presence of the STK11 mutation suggests a potentially diminished efficacy and a higher likelihood of resistance mechanisms emerging. Considering the options, a KRAS G12C inhibitor is the most appropriate targeted therapy. However, the question asks for the *most likely* outcome considering the molecular context. The combination of KRAS G12C and STK11 mutations often predicts a less robust response to KRAS inhibitors, and the patient has already progressed on immunotherapy, suggesting a limited benefit from further immune checkpoint blockade alone. Therefore, the most accurate prediction is a limited duration of response.
-
Question 30 of 30
30. Question
A 68-year-old male with a history of heavy smoking presents with newly diagnosed metastatic non-small cell lung cancer (NSCLC). Molecular profiling reveals no actionable driver mutations. He is initiated on pembrolizumab, a PD-1 inhibitor, as first-line therapy. Following six cycles, imaging demonstrates progressive disease. Biopsy of a metastatic lesion reveals significant infiltration of CD8+ T cells within the tumor parenchyma, but these T cells exhibit limited cytotoxic activity and are surrounded by a dense population of myeloid cells expressing arginase-1. Considering the complex interplay within the tumor microenvironment and its impact on immunotherapy response, which of the following cellular components within the tumor microenvironment is most likely contributing to the observed resistance to PD-1 blockade in this patient?
Correct
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immune checkpoint inhibitors (ICIs). The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) exhibiting a poor response to PD-1 blockade. The key to answering this question lies in understanding the multifaceted nature of the TME and how its components can either synergize with or antagonize anti-tumor immunity. A critical factor influencing ICI response is the presence and function of myeloid-derived suppressor cells (MDSCs). MDSCs are a heterogeneous population of immature myeloid cells that accumulate in the TME and actively suppress T cell responses through various mechanisms, including the production of immunosuppressive cytokines (e.g., IL-10, TGF-β) and the expression of enzymes like arginase-1 and inducible nitric oxide synthase (iNOS). These suppressive functions directly impair the activation, proliferation, and effector functions of cytotoxic T lymphocytes (CTLs), which are essential for ICI efficacy. In this scenario, the observed lack of response to PD-1 blockade, despite initial tumor infiltration by CD8+ T cells (indicating some level of immune recognition), strongly suggests an overriding immunosuppressive mechanism within the TME. The presence of a high density of MDSCs, particularly those expressing arginase-1, would effectively dampen the anti-tumor activity of the CD8+ T cells, rendering them unable to overcome the PD-1 mediated inhibition. Therefore, targeting MDSCs, for instance, by inhibiting their suppressive functions or promoting their differentiation into less immunosuppressive myeloid cells, would be a logical strategy to enhance ICI efficacy. Other factors, while relevant to the TME, are less directly implicated in explaining the specific pattern of poor response despite CD8+ infiltration. For example, while tumor mutational burden (TMB) is a known predictor of ICI response, the question implies that the immune system is initially engaged. Similarly, tumor-associated macrophages (TAMs) can have diverse roles, but a predominant M2-like phenotype (often associated with immunosuppression) would also contribute to resistance, but MDSCs are a more direct and potent suppressors of T cell function in many contexts. Altered tumor cell metabolism can impact T cell function, but it’s a less direct explanation for the observed resistance pattern compared to direct immune suppression by cellular components of the TME. Therefore, the most likely explanation for the patient’s non-response, given the provided information, is the presence of a highly immunosuppressive TME dominated by MDSCs that are actively inhibiting T cell effector functions.
Incorrect
The question probes the understanding of tumor microenvironment (TME) interactions and their impact on therapeutic efficacy, specifically in the context of immune checkpoint inhibitors (ICIs). The scenario describes a patient with advanced non-small cell lung cancer (NSCLC) exhibiting a poor response to PD-1 blockade. The key to answering this question lies in understanding the multifaceted nature of the TME and how its components can either synergize with or antagonize anti-tumor immunity. A critical factor influencing ICI response is the presence and function of myeloid-derived suppressor cells (MDSCs). MDSCs are a heterogeneous population of immature myeloid cells that accumulate in the TME and actively suppress T cell responses through various mechanisms, including the production of immunosuppressive cytokines (e.g., IL-10, TGF-β) and the expression of enzymes like arginase-1 and inducible nitric oxide synthase (iNOS). These suppressive functions directly impair the activation, proliferation, and effector functions of cytotoxic T lymphocytes (CTLs), which are essential for ICI efficacy. In this scenario, the observed lack of response to PD-1 blockade, despite initial tumor infiltration by CD8+ T cells (indicating some level of immune recognition), strongly suggests an overriding immunosuppressive mechanism within the TME. The presence of a high density of MDSCs, particularly those expressing arginase-1, would effectively dampen the anti-tumor activity of the CD8+ T cells, rendering them unable to overcome the PD-1 mediated inhibition. Therefore, targeting MDSCs, for instance, by inhibiting their suppressive functions or promoting their differentiation into less immunosuppressive myeloid cells, would be a logical strategy to enhance ICI efficacy. Other factors, while relevant to the TME, are less directly implicated in explaining the specific pattern of poor response despite CD8+ infiltration. For example, while tumor mutational burden (TMB) is a known predictor of ICI response, the question implies that the immune system is initially engaged. Similarly, tumor-associated macrophages (TAMs) can have diverse roles, but a predominant M2-like phenotype (often associated with immunosuppression) would also contribute to resistance, but MDSCs are a more direct and potent suppressors of T cell function in many contexts. Altered tumor cell metabolism can impact T cell function, but it’s a less direct explanation for the observed resistance pattern compared to direct immune suppression by cellular components of the TME. Therefore, the most likely explanation for the patient’s non-response, given the provided information, is the presence of a highly immunosuppressive TME dominated by MDSCs that are actively inhibiting T cell effector functions.