Skip to main content
Special Report

2024 NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) Update: Impact on NSCLC Landscape

11/15/2024

The updated 2024 NCCN Guidelines® recommend broad genomic testing and the need for multidisciplinary care to accurately diagnose and treat non-small cell lung cancer. View this special report to learn more.  

NSCLC DISEASE PREVALENCE AND BURDEN

Lung cancer, though constituting only approximately 13% of all cancers, is the leading cause of cancer-related deaths in the United States and worldwide.1,2 In 2021, approximately 469 000 new cases of lung and bronchus cancer were diagnosed in the United States.3 While incidence and mortality have decreased over the past several years, lung cancer still accounts for more deaths than colorectal, breast, brain, and prostate cancers, with approximately 340 people dying from lung cancer every day.4,5 The 5-year lung cancer survival rates remain one of the lowest of all cancer types, at only 25%.5

Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and accounts for approximately 85% of all lung cancer cases.1 Traditionally, patients with advanced NSCLC have had limited treatment options. Over the past 2 decades, however, a variety of tumor-specific genetic aberrations have been discovered and used to predict prognosis and response to targeted therapies, particularly for patients with adenocarcinoma.6,7

Patients with NSCLC often face significant declines in their health-related quality of life due to both disease progression and adverse effects associated with treatment. They can experience a wide range of symptoms, including loss of appetite, pain, dyspnea, fatigue, cough, and blood in the sputum. In addition to these physical effects, many patients will be forced to reduce their working hours in order to receive treatment and attend medical appointments, which can interrupt normalcy in their daily lives and deter career advancement.8

In recent years, survival rates in patients with NSCLC have improved largely due to better disease management, availability of new treatments including immunotherapy and targeted therapy, earlier detection, and advances in staging.1,5 The 3-year survival rate of NSCLC has increased from 25% to 38% since 2000.4 Ongoing research continues to focus on improving survival rates for patients with NSCLC.

TARGETED THERAPIES IN NSCLC:  BACKGROUND AND EVOLUTION

Tailoring treatments to subtypes of NSCLC (adenocarcinoma, squamous cell carcinoma, and large cell carcinoma) was a relatively new concept 20 years ago.9,10 The platinum-based doublet chemotherapy, a standard of care for treating advanced NSCLC, reached an efficacy plateau over time. With genesis in other tumor types, the idea of targeting the driver mutations of a particular tumor to inhibit cell proliferation and survival began to take hold in NSCLC as well.11  

The first targetable mutations discovered in lung cancer were epidermal growth factor receptor (EGFR) mutations. NSCLC with an overexpression of EGFR is known to be associated with an increased risk of metastasis, a high rate of tumor growth, and poor tumor differentiation.7 Tyrosine kinase inhibitors (TKIs) were initially developed to target EGFR, with the first dosing study of gefitinib occurring in 1998. Significant improvements in outcomes were observed throughout the gefitinib trials, particularly in female patients, patients of Asian origin, patients with adenocarcinoma, and never-smokers.12 Over time, more EGFR mutations have been identified, including in-frame deletions of exon 19, exon 20 insertions, and L858R substitutions in exon 21.7,13 While EGFR remains one of the most significant driver mutations in NSCLC patients, other driver mutations within NSCLC to potentially target include ALK, ROS1, BRAF, KRAS, MET, RET, NTRK, and HER2 (Table 1).7

Therapies are continuously being developed to target these genetic aberrations, and guidelines are constantly being updated.1,6 As an example, in August 2024, the Food and Drug Administration (FDA) approved lazertinib in combination with amivantamab-vmjw for the first-line treatment of locally advanced or metastatic NSCLC with EGFR exon 19 deletions or exon 21 L858R substitution mutations.19 One of the most recent updates to the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for NSCLC, published less than a month after this approval, reflects the use of new targeted therapies in NSCLC patients, including amivantamab-vmjw + lazertinib as a treatment option, to help achieve better outcomes.16 

Table 1

 

AN OVERVIEW OF THE 2024 NSCLC NCCN GUIDELINES® UPDATES

The NCCN Guidelines for NSCLC were updated a number of times in 2024, with the most recent iteration, Version 10.2024, published on September 23, 2024. This Special Report summarizes the guidelines changes that occurred in the past year. The guidelines now highlight advanced testing procedures and changes in treatment pathways for NSCLC depending on clinical presentation and biomarkers present.16

Principles of Diagnostic Evaluation

Recent updates to the NCCN Guidelines emphasize multidisciplinary collaboration when diagnosing the disease, including the involvement of both treating physicians and specialists in obtaining tissue diagnosis prior to treatment.16

The approach to pursuing a preoperative biopsy was further explained. The decision needs to be informed by the pretest probability of malignancy, including consideration of radiologic appearance, risk factors, and residence in areas with widespread endemic infectious lung disease. Overall, the updated guidelines state that a preoperative biopsy may be an option for several situations, including a non-lung cancer diagnosis (ie, granulomatous nodule from endemic fungus), suspected stage IB+ lung cancer in patients who may be eligible for systemic therapy prior to surgery, an intraoperative diagnosis that may be challenging or risky to complete, or to establish a diagnosis prior to beginning stereotactic ablative radiotherapy (SABR) in patients.16 

Diagnostic tools and methods for tissue diagnosis were expanded upon, with the addition of left anterior mediastinotomy/chamberlain as a technique that should routinely be available. Guidelines now include robotic navigational bronchoscopy as an option for patients with pulmonary nodules, in addition to the standard navigational bronchoscopy, radial endobronchial ultrasound (EBUS), or transthoracic needle aspiration.16

Image
Table 2

Principles of Molecular and Biomarker Analysis

Recent updates place an increased emphasis on biomarker testing, recommending plasma or biopsy testing for EGFR, KRAS, ALK, ROS1, BRAF, NTRK1/2/3, MET, RET, and ERBB2 (HER2). Even if rapid testing is done, broad-based testing should also be completed with combinations of tissue and plasma testing, concurrently if possible. If results are negative, guidelines recommend retesting with a complementary method. If an assay has a technical failure, consider an alternative testing modality or procure an additional tissue sample to achieve broad molecular profiling. Once a clinically actionable marker is found, targeted therapy for that marker should be started, if available.16  

There are several limitations with both circulating tumor DNA (ctDNA) and tissue testing, which are described in Table 2. Guidelines now state that ctDNA is not routinely recommended in settings other than advanced/metastatic disease. For stages I to III, tissue-based testing is preferred, with metastatic disease confined to the thorax. This potentially results in a higher yield with tissue-based testing. Because both ctDNA and tissue testing have significant false-negative rates, complementary testing is recommended to shorten test yield time while improving the identification of treatment targets.16 

Mediastinal Biopsy Categories

Mediastinal biopsy categories have now been modified into 4 categories. Categories of operable and medically inoperable were added to all except T1-3, N2 nodes positive, and M0.16

Pretreatment Evaluation

The definition of “medically inoperable” was expanded upon, now including high-risk patients as determined by a thoracic surgeon or those who decline surgery after thoracic surgical consultation.16  

Additional modifications were made for the pretreatment evaluation for patients in stage IB-IIIA at clinical assessment. In this patient population, negative mediastinal nodes are now classified as “no nodal disease.” Recommended regimens for initial treatment remain the same, except that adjuvant chemotherapy can now be considered for high-risk stage II instead of high-risk stage IB disease. Positive mediastinal nodes were changed to “N1 or N2 disease,” with initial treatment options remaining the same except for the addition of stage IIB treatment.16

Principles of Surgical Therapy - Resection

For chest wall, trachea/carina, or mediastinum; T3 invasion, N0-1; resectable T4 extension, N0-1; and stage IIIA (T4, N0-1) resectable clinical presentations, initial treatment options were modified. Surgery remains the preferred treatment option, but guidelines removed “preoperative systemic therapy if planned” and added the footnote “resectability should be determined by thoracic surgery evaluation prior to initiation of any therapy.”16

For sublobar resection, segmentectomy and wedge resection should now be strongly considered for peripheral T1ab, N0 tumors. Segmentectomy and wedge resection are noted to be appropriate for patients with poor pulmonary reserve or other significant comorbidities that contraindicate lobectomy.16

In the case of robotic surgery, additional context was added with the guidelines citing that studies of robotic-assisted pulmonary resection have shown that this method is not inferior to traditional video-assisted thoracic surgery (VATS) approaches when performed by experienced robotic surgeons.16

Principles of Surgical Therapy - Margins and Nodal Assessment

A bullet on N1 and N2 node resection and mapping was modified, stating that a minimum of 1 N1 and 3 N2 stations are needed to be sampled or lymph node dissection should be completed. Formal ipsilateral mediastinal lymph node dissection was initially indicated for patients undergoing resection for stage IIIA disease but is now indicated for N2 disease instead. Guidelines still recommend considering referral to a radiation oncologist in these cases.16

Perioperative Systemic Therapy

For patients being evaluated for neoadjuvant systemic therapy, the guidelines have expanded the consideration to cover the whole immune checkpoint inhibitor (ICI) class (unless contraindicated) + chemotherapy for those with tumor size ≥ 4 cm or are node-positive. Alternatively, patients who are ineligible for ICI can be treated with induction systemic therapy after surgical evaluation. Testing recommendations now include EGFR, ALK rearrangements, and PD-L1 status.16

For patients who are candidates for ICIs without EGFR mutations or ALK rearrangements, a new regimen was added consisting of durvalumab 1500 mg and platinum-based doublet chemotherapy every 3 weeks for 4 cycles. Platinum-doublet chemotherapy options were also added (Table 3). Durvalumab may then be continued as a single-agent adjuvant treatment after surgery.16

For systemic therapy following surgical resection, a new NCCN Category 1 regimen of durvalumab 1500 mg every 4 weeks for up to 12 cycles was added as a treatment option. This regimen is for patients with node-positive NSCLC and/ or completely resected tumors ≥ 4 cm who have previously received neoadjuvant durvalumab + chemotherapy. Again, this regimen is for patients without an EGFR mutation or ALK rearrangement.16 

Image
Table 3

Principles of Radiation Therapy

Additional evidence was added for intensity-modulated proton therapy (IMPT), including a series of retrospective studies in which IMPT has been shown to reduce toxicity compared with 3D-based passive scattering proton therapy in stage III NSCLC.16

 For early-stage NSCLC (stage I, selected node-negative stage IIA), studies have shown that SABR, also known as stereotactic body radiation therapy (SBRT), has demonstrated similar overall and cancer-specific survival compared to lobectomy while showing reduced acute toxicity. Therefore, it is now deemed an appropriate option for patients with high surgical risk due to poor cardiac function, in addition to characteristics such as age ≥ 75 years or poor lung function, which already were included in the guidelines.16

Image-guided thermal ablation (IGTA) therapy is now an option for select patients regardless of whether they previously received SABR, definitive radiation therapy (RT) SABR, or definitive RT.16  

The guidelines exchanged 3D-conformal radiation therapy (3D-CRT) regimens for highly conformal radiation (intensity-modulated radiation therapy, with image-guided radiation therapy preferred) as less preferred alternatives to consider if referral for SABR is not feasible.16  

For locally advanced NSCLC (Stage II-III), in patients with completely resected pN1 receiving adjuvant systemic therapy, the use of postoperative radiotherapy (PORT) is not recommended; PORT may be considered for these patients if they cannot receive adjuvant systemic therapy.16

For palliative RT for advanced/metastatic NSCLC, SABR stereotactic radiosurgery (SRS) has been shown in randomized clinical trials to achieve better pain and tumor control of bone metastases  (spine and non-spine) than conventional RT. This strategy may be appropriate for patients with better survival prognoses.16  

Pharmacological Treatment Updates

Adjuvant treatment was adjusted for separate pulmonary nodule(s), same lobe (T3, N0-1), or ipsilateral non-primary lobe (T4, N0-1) with negative margins after surgery. Previously, treatment options included chemotherapy as Category 1 or the option of sequential chemotherapy. This has been replaced with adjuvant systemic therapy or sequential chemotherapy and the consideration of RT.16

Treatment Updates by Biomarkers Present

Recent NCCN Guidelines updates modified the treatment pathways for first-line and subsequent therapies based on mutations present, past treatments, and potential resistance. Repotrectinib, amivantamab, osimertinib, durvalumab, and lorlatinib were highlighted often as new updates.16 

The updated guidelines for subsequent systemic therapy for advanced or metastatic disease note that “some oncogenic drivers (ie, EGFR exon 19 deletion or exon 21 L858R, ALK rearrangements) are associated with less benefit from PD-1/PD-L1 inhibitors.” Updates also warn of the potential for toxicity when administering a TKI in temporal proximity to immunotherapy in patients who are positive for EGFR exon 19 deletion or exon 21 L858R.16

EGFR

For patients with an EGFR exon 19 deletion or exon 21 L858R mutation discovered prior to first-line therapy, osimertinib is a preferred treatment option. Amivantamabvmjw + lazertinib and osimertinib + pemetrexed + (cisplatin or carboplatin) (nonsquamous) have been added as “Other Recommended” options for first-line therapy (Category 1 recommendations). Additionally, the following regimens are now classified as “Useful in Certain Circumstances,” instead of “Other Recommended”: erlotinib + ramucirumab, erlotinib + bevacizumab, dacomitinib (Category 1), afatinib (Category 1), erlotinib (Category 1), or gefitinib (Category 1).16  

If EGFR exon 19 deletion or exon 21 L858R mutations are discovered during first-line systemic therapy, the recommendation is to complete planned therapy or interrupt the therapy followed by the option of treatment with osimertinib (preferred) or amivantamab-vmjw + lazertinib. A footnote regarding the use of osimertinib in combination or following immune checkpoint inhibitors was modified to include “the rate of side effects (pneumonitis) is higher within 3 months.” For amivantamab-vmjw + lazertinib use, a footnote was added to recommend prophylactic anticoagulation at treatment initiation to prevent venous thromboembolism events.16

Adjuvant treatment was modified for patients who have received sequential chemoradiation, now stating that durvalumab can be considered as consolidation immunotherapy (Category 1) or, if EGFR exon 19 deletion or exon 21 L858R is present, osimertinib (Category 1) is recommended as a treatment option. This update was included for treatment after the following clinical presentations of disease: superior sulcus tumor (T4 extension, N0-1), stage IIIA (T4, N0-1) unresectable, T1-3 N1 nodes positive M0, T1-3 N2 nodes positive M0, stage IIIB (T1-2, N3), stage IIIC (T3, N3), stage IIIB (T4, N2), and stage IIIC (T4, N3).16

For systemic therapy following surgical resection of stage IB-IIIA or stage IIIB (T3, N2) in patients with EGFR exon 19 deletion or exon 21 L858R who have received previous adjuvant chemotherapy or are ineligible to receive platinum-based chemotherapy, guidelines also have extended the treatment dosage for osimertinib to 3 years.16

Additionally, for patients with the EGFR exon 19 deletion or exon 21 L858R mutation, amivantamab-vmjw + carboplatin + pemetrexed has been added as an option for subsequent therapy (nonsquamous histology), if an amivantamab-vmjwN-based regimen was not previously given. It is also a Category 1, Preferred regimen for patients with the EGFR exon 19 deletion or exon 21 L858R mutation (nonsquamous histology) who have systemically progressed on osimertinib and are symptomatic, with multiple lesions. However, guidelines modified a footnote to highlight the consideration of “biopsy or plasma testing to evaluate mechanisms of resistance.” For asymptomatic or symptomatic progression with brain or limited systemic disease, continuation of amivantamab-vmjw + lazertinib was added as a treatment option.16

The amivantamab-vmjw + carboplatin + pemetrexed regimen is now a Category 1, Preferred option for first-line treatment of patients with nonsquamous EGFR exon 20 insertion mutation. For patients with an EGFR S768I, L861Q, and/or G719X mutation, amivantamab-vmjw + carboplatin + pemetrexed was also added as an option for subsequent therapy of advanced or metastatic nonsquamous disease.16

Finally, guidelines cite that in patients with EGFR-mutant NSCLC who develop high-level MET amplifications, administration of capmatinib, tepotinib, or crizotinib with continuation of osimertinib is acceptable.16 

ALK Rearrangements

For patients with resistant mutations, including ALK G1202R and L1196M (except compound L1196M/G1202R), lorlatinib is now an option for therapy after progression.16

For systemic therapy following surgical resection, guidelines have added alectinib 600 mg twice daily for 24 months as a Category 1 treatment option for patients with completely resected stage II to IIIA or stage IIIB (T3, N2) NSCLC and positive for ALK rearrangements.16

ROS1

Repotrectinib was added as a preferred first-line treatment for patients with ROS1 rearrangements and as an option alongside entrectinib for patients with brain metastases. For subsequent treatment of asymptomatic disease, repotrectinib was added as an option, if not previously given. For subsequent treatment of symptomatic brain disease, options include entrectinib (if previously treated with crizotinib or ceritinib), repotrectinib (if previously treated with crizotinib, ceritinib, or entrectinib), or lorlatinib. Finally, for symptomatic, systemic disease with multiple lesions, newly added options include repotrectinib, if not previously given, or lorlatinib.16

NTRK Fusions

For patients with NTRK fusions, repotrectinib is now a preferred f irst-line treatment option if discovered prior to beginning f irst-line systemic therapy. If discovered during first-line systemic therapy, guidelines recommend continuing planned systemic therapy (including maintenance) or allowing therapy to be interrupted and followed by larotrectinib, entrectinib, or repotrectinib. Repotrectinib was added as a treatment option for subsequent therapy, if not previously given.16  

PD-L1

For patients expressing PD-L1 ≥ 1% to 49%, cemiplimab-rwlc + pemetrexed + (carboplatin or cisplatin) is now a Category 1, Preferred treatment option for first-line therapy for advanced or metastatic disease of adenocarcinoma, large cell, NSCLC  (PS 0-2). Additionally, cemiplimab-rwlc + paclitaxel + (carboplatin or cisplatin) is now a Category 1, Preferred treatment option for first-line therapy of squamous cell carcinoma (PS 0-2).16

For patients expressing PD-L1 ≥ 50%, cemiplimab-rwlc + pemetrexed + (carboplatin or cisplatin) is now a Category 1, Preferred treatment option for first-line therapy for advanced or metastatic disease of adenocarcinoma, large cell, NSCLC not otherwise specified (NOS) (PS 0-2), and cemiplimab-rwlc + paclitaxel + (carboplatin or cisplatin) is now a Category 1, Preferred treatment option for first-line therapy of squamous cell carcinoma (PS 0-2).16

In addition, cemiplimab-rwlc + pemetrexed + (carboplatin or cisplatin) was added as a Category 1, Preferred treatment option for systemic therapy of advanced or metastatic disease of adenocarcinoma, large cell, NSCLC NOS (PS 0-1) and squamous cell carcinoma (PS 0-1), with no contraindications to PD-1 or PD-L1 inhibitors.16 Atezolizumab monotherapy is an option for patients with PS 3, regardless of PD-L1 status.

Atezolizumab intravenous infusion and atezolizumab + hyaluronidase-tqjs subcutaneous injection can now both be considered, noting differences in dosing and administration schedules between the 2 modes of administration.16 

HER2

Fam-trastuzumab deruxtecan-nxki was added as an “Other Recommended” option for subsequent therapy of PS 0-2 for adenocarcinoma, large cell, NSCLC NOS, and squamous cell carcinoma (PS 0-2), regardless of previous immuno-oncology therapy. Fam-trastuzumab deruxtecan-nxki was also added as a treatment option for progression therapy of PS 0-2 adenocarcinoma, large cell, NSCLC NOS, and squamous cell carcinoma. However, a footnote was added to ensure this treatment is only used for tumors with HER2 overexpression (IHC 3+).16 

DISCUSSION: IMPACT OF THE GUIDELINES UPDATES ON THE NSCLC LANDSCAPE

Broad Molecular & Biomarker Testing and Targeted Therapy

The NCCN Guidelines updates highlight the importance of biomarker testing prior to treatment initiation, supporting the importance of personalized treatment to circumvent resistance, prevent unnecessary toxicity, and improve outcomes.1,6,16

Targeted therapy has greatly improved patient outcomes in NSCLC and other tumors, particularly when treating advanced tumors unresponsive to conventional therapies.1 Because some of the aforementioned oncogenic driver mutations are uncommon, testing for these mutations does not routinely occur due to cost, time, and/or limited size of tumor samples for diagnosis.6

The 2024 NCCN Guidelines updates place an increased emphasis on broad molecular testing for these oncogenic drivers. Recommending full testing of all potential biomarkers will facilitate appropriate treatment earlier in patients. Additionally, guidelines describe the limitations of tissue and ctDNA testing, particularly the significant number of false-negative results. Because of this, complementary testing is recommended to ensure that the presence of genomic alterations is accurately captured.16 A recent study suggested that the complementary approach of ctDNA and tissue testing after lung cancer diagnosis increases the number of patients receiving broad molecular testing and thus the delivery of targeted therapies to advanced NSCLC patients. This study also found that plasma ctDNA testing resulted in more frequent identification of actionable mutations in comparison to tissue testing alone. Complementary ctDNA and tissue testing could potentially allow patients to start targeted therapies earlier, resulting in better outcomes.20

The updated guidelines include options for different therapies to begin depending on the biomarker(s) present, the line of therapy, and the prior therapy given. Therapies such as osimertinib, amivantamab, repotrectinib, and lorlatinib, among others, were highlighted in the recent updates.16 For those patients without an actionable mutation, combination chemotherapy is the standard regimen, with a median overall survival for advanced or metastatic disease of less than 2 years.21 Clear recommendations on which therapies to use in the presence of targetable mutations will allow healthcare professionals to provide the most effective therapy to their patients, potentially increasing response rates and extending survival.7,21

Determining which treatments are most effective in the firstline setting is essential, as this decision also establishes subsequent lines of therapy upon treatment failure. Understanding the presence of oncogenic drivers can provide additional, more personalized options for the treatment of NSCLC.6

Diagnosis and Evaluation

The NCCN Guidelines updates outlined when to perform a preoperative biopsy, noting the time, cost, and risks associated with this procedure. More diagnostic tools were recommended, with the inclusion of robotics in certain situations, highlighting an increase in the usage of artificial intelligence within healthcare.16,22 Mediastinal lymph node involvement is the second most meaningful predictor of curative survival for NSCLC patients, behind the presence or absence of distant metastases.4

Mediastinal biopsy categories were modified in the 2024 updates.16 Determining which lymph node stations are involved with metastatic spread is essential when determining the proper stage, prognosis, and appropriate therapy for patients. Accurate staging results in improved survival.4  

Patient outcomes can be improved by adding clarity to the staging and selection of the appropriate treatment regimen. 

Treatment Landscape

A common theme in all guidelines updates is the timely evaluation and incorporation of new evidence to support NSCLC management.16 The aforementioned approval of lazertinib in combination with amivantamab-vmjw expands the treatment armamentarium of NSCLC. Efficacy and safety data of amivantamab-vmjw + lazertinib were evaluated in the MARIPOSA trial, a randomized, active-controlled, multicenter trial consisting of 1074 patients with EGFR exon 19 deletion or exon 21 L858R substitution mutations, locally advanced or metastatic NSCLC, with no prior systemic therapy for advanced disease. The median PFS was 23.7 months compared to 16.6 months in the comparator arm (osimertinib). The most common adverse reactions, occurring in ≥ 20% of patients, included rash, nail toxicity, infusion-related reactions, musculoskeletal pain, edema, stomatitis, venous thromboembolism, paresthesia, fatigue, diarrhea, constipation, COVID-19 infection, hemorrhage, dry skin, decreased appetite, pruritus, nausea, and ocular toxicity. Due to the seriousness of venous thromboembolism, it is recommended that patients taking this regimen should also take a prophylactic anticoagulation for the first 4 months of therapy.19,23 The approval of treatments such as this, followed by inclusion within the NCCN Guidelines, allows healthcare providers more options when selecting the most optimal treatment for their patients.

The number of updates in 2024 reflects how quickly the NSCLC landscape can change and the importance of staying abreast of research, as it holds the key to better understanding how best to treat patients and achieve positive outcomes. 

Multidisciplinary Collaboration

Interdisciplinary collaboration is deemed essential in correctly diagnosing lung cancer, with guidelines recommending that both treating physicians and specialists in obtaining tissue diagnosis (thoracic surgery, interventional pulmonology, and interventional radiology) be involved in the decision-making process from the beginning.16 Studies have shown that multidisciplinary care in lung cancer has been associated with improved patient outcomes, including survival, greater utilization of appropriate treatment modalities, and a better alignment of treatment recommendations and management guidelines.24 Multidisciplinary care is also advocated for by both the American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO).25 

CONCLUSION

Lung cancer remains the leading cause of cancer-related death worldwide.5 NSCLC, the most common type of lung cancer, has seen significant improvements in outcomes over the past 2 decades with the advent of immunotherapy and targeted therapies, better disease management, advancements in staging, and earlier detection of disease.1,5 Echoing the increasing importance of identifying and targeting actionable biomarkers, the 2024 NCCN Guidelines updates recommend broad genomic testing and the need for multidisciplinary care for the accurate diagnosis and treatment of NSCLC. Staging categories were modified, underscoring the relationship between proper staging and prognosis. Additionally, principles of diagnostic and pretreatment evaluation, surgical resection, and radiation therapy were all updated over the past year. The guidelines added key details on 8 new targeted therapeutic options depending on which genetic aberrations may be present. Focusing on delivering the right treatment to the right patient is of utmost importance and is reflected in these updates.16

Disclaimer: NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way.

Medical writing assistance was provided by HMP Collective.

REFERENCES

  1. Araghi M, Mannani R, Heidarnejad Maleki A, et al. Recent advances in non-small cell lung cancer targeted therapy; an update review. Cancer Cell Int. 2023;23(1):162. doi:10.1186/s12935-023-02990-y
  2. Ganti AK, Klein AB, Cotarla I, Seal B, Chou E. Update of incidence, prevalence, survival, and initial treatment in patients with non–small cell lung cancer in the US. JAMA Oncol. 2021;7(12):1824. doi:10.1001/jamaoncol.2021.4932
  3. SEER*Explorer: an interactive website for SEER cancer statistics. Surveillance Research Program, National Cancer Institute. Updated June 27, 2024. Accessed September 12, 2024. https://seer.cancer.gov/statistics-network/explorer/
  4. Rodriguez GR, Trachiotis GD, Mullenix PS, Antevil JL. Minimally invasive with maximal yield: a narrative review of current practices in mediastinal lymph node staging in non-small cell lung cancer. J Laparoendosc Adv Surg Tech. Published online May 10, 2024. doi:10.1089/lap.2024.0138
  5. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74(1):12-49. doi:10.3322/caac.21820
  6. Herrera-Juárez M, Serrano-Gómez C, Bote-de-Cabo H, Paz-Ares L. Targeted therapy for lung cancer: beyond EGFR and ALK. Cancer. 2023;129(12):1803-1820. doi:10.1002/cncr.34757
  7. Melosky B, Kambartel K, Häntschel M, et al. Worldwide prevalence of epidermal growth factor receptor mutations in non-small cell lung cancer: a meta-analysis. Mol Diagn Ther. 2022;26(1):7-18. doi:10.1007/s40291-021-00563-1
  8. Tufman A, Redmond K, Giannopoulou A, et al. Patient perception of burden of disease and treatment preferences in non-small cell lung cancer: results from a European survey. Lung Cancer. 2022;168:59-66. doi:10.1016/j.lungcan.2022.04.008
  9. Cooper WA, O’Toole S, Boyer M, Horvath L, Mahar A. What’s new in non-small cell lung cancer for pathologists: the importance of accurate subtyping, EGFR mutations and ALK rearrangements. Pathology (Phila). 2011;43(2):103-115. doi:10.1097/ PAT.0b013e328342629d
  10. Chan BA, Hughes BGM. Targeted therapy for non-small cell lung cancer: current standards and the promise of the future. Transl Lung Cancer Res. 2015;4(1):36-54. doi:10.3978/j.issn.2218-6751.2014.05.01
  11. Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011;12(2):175-180. doi:10.1016/S1470-2045(10)70087-5
  12. Vansteenkiste J, Wauters E. Tyrosine kinase inhibition of EGFR: a successful history of targeted therapy for NSCLC since 20 years. Ann Oncol. 2018;29:i1-i2. doi:10.1093/annonc/mdx724
  13. Burnett H, Emich H, Carroll C, Stapleton N, Mahadevia P, Li T. Epidemiological and clinical burden of EGFR exon 20 insertion in advanced non-small cell lung cancer: a systematic literature review. PLoS ONE. 2021;16(3):e0247620. doi:10.1371/journal.pone.0247620
  14. Brazel D, Nagasaka M. The development of amivantamab for the treatment of nonsmall cell lung cancer. Respir Res. 2023;24(1):256. doi:10.1186/s12931-023-02558-4
  15. Muminovic M, Carracedo Uribe CR, Alvarez-Pinzon A, Shan K, Raez LE. Importance of ROS1 gene fusions in non-small cell lung cancer. Cancer Drug Resist. 2023;6(2):332-344. doi:10.20517/cdr.2022.105
  16. Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Non-Small Cell Lung Cancer V.10.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed September 29, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org.
  17. Azelby CM, Sakamoto MR, Bowles DW. ROS1 targeted therapies: current status. Curr Oncol Rep. 2021;23(8):94. doi:10.1007/s11912-021-01078-y
  18. Shih JY. ERBB2 amplification in NSCLC: how many faces? J Thorac Oncol. 2024;19(5):668-670. doi:10.1016/j.jtho.2024.02.001
  19. FDA approves lazertinib with amivantamab-vmjw for non-small lung cancer. US Food and Drug Administration. August 19, 2024. Accessed August 30, 2024. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approveslazertinib-amivantamab-vmjw-non-small-lung-cancer
  20. García-Pardo M, Czarnecka-Kujawa K, Law JH, et al. Association of circulating tumor DNA testing before tissue diagnosis with time to treatment among patients with suspected advanced lung cancer: the ACCELERATE nonrandomized clinical trial. JAMA Netw Open. 2023;6(7):e2325332. doi:10.1001/jamanetworkopen.2023.25332
  21. Wang M, Herbst RS, Boshoff C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat Med. 2021;27(8):1345-1356. doi:10.1038/s41591021-01450-2
  22. Kumar KS, Miskovic V, Blasiak A, et al. Artificial intelligence in clinical oncology: from data to digital pathology and treatment. Am Soc Clin Oncol Educ Book. 2023;(43):e390084. doi:10.1200/EDBK_390084
  23. Cho BC, Lu S, Felip E, et al. Amivantamab plus lazertinib in previously untreated EGFR-mutated advanced NSCLC. N Engl J Med. Published online June 26, 2024. doi:10.1056/NEJMoa2403614
  24. Heinke MY, Vinod SK. A review on the impact of lung cancer multidisciplinary care on patient outcomes. Transl Lung Cancer Res. 2020;9(4):1639-1653. doi:10.21037/ tlcr.2019.11.03
  25. Morabito A, Mercadante E, Muto P, et al. Improving the quality of patient care in lung cancer: key factors for successful multidisciplinary team working. Explor Target Anti-Tumor Ther. 2024;5(2):260-277. doi:10.37349/etat.2024.00217