Lung


Presentation

Resectability

Recommended treatment

T1-2N0

Operable

Lobectomy (preferred over segmentectomy or wedge resection) or SBRT

Inoperable

SBRT (may consider RFA/Cryotherapy)

II (T2bN0, T1-2N1, T3N0)

Operable

Surgery → chemo (>4 cm)

Inoperable

ChemoRT → ±chemo or hypofx EBRT → ±chemo

IIIA

Operable

ChemoRT → restage → surgery → chemo or Chemo → restage → surgery → chemo ± RT

Inoperable

ChemoRT → ±chemo

IIIB

Inoperable

ChemoRT → ±chemo

Recurrent

Operable

EBRT/SBRT/resection for limited local recurrence → systemic therapy

Inoperable

EBRT/SBRT/RFA/cryo for limited recurrence → systemic therapy

Pulmonary oligometastases

Operable

Lobectomy/wedge resection or SBRT or hypofractionated EBRT (for larger lesions, >5 cm) → systemic therapy

Inoperable

SBRT, RFA, cryo, or hypofx EBRT (preferred for larger lesions, > 5 cm) → systemic therapy








    Radiosurgical Technique



    Simulation and Treatment Planning






    • Tumor motion may be 2–3 cm in peri-diaphragmatic regions of the lower lung. Motion management strategies include respiratory gating , coaching with audio-visual feedback, breath-hold techniques, abdominal compression , and intrafraction tumor tracking real-time imaging techniques with dynamic beam and/or couch compensation.


    • Thin-cut CT (≤1.5 mm) thickness recommended. 4DCT or maximal inspiratory and expiratory phase CTs or slow CT recommended to assess target and critical structure internal motion. Free-breathing helical or mean intensity projection CT should be used for dose calculation.


    • iGTV contoured from Maximum Intensity Projection (MIP) generated from 4DCT. MIP should be used judiciously in tumors adjacent to diaphragm or chest wall , with additional imaging as needed to fully discriminate the target from surrounding normal tissue with similar CT tissue density.


    • GTV /iGTV  = tumor visible on CT lung window.


    • CTV /ITV  = GTV /iGTV  + 0–10 mm (in RTOG protocols , GTV and CTV have been considered identical on CT planning with zero expansion margin added).


    • PTV  = CTV /ITV  + 3–10 mm (dependent upon available center-specific IGRT and motion management capabilities). Current RTOG guidelines are:



      • Non-4DCT planning, PTV  = GTV  + 5 mm axial and 10 mm longitudinal anisotropic margins.


      • 4DCT planning, PTV  = ITV  + 5 mm isotropic margin.


    • Dose to proximal OARs attributed to compact intermediate dose region outside of the CTV /ITV region, generally reduced with increased beams and angles, as well as minimization of margins on target.


    • Treatment planning guidelines (adapted from RTOG 0618).



      • VRx dose ≥95 % PTV , V90 ≥99 % PTV.


      • High dose region (≥105 % Rx dose) should fall within the PTV .


      • Conformality Index goal ≤1.2.


    • Heterogeneity correction algorithms are increasingly routinely used for planning (anisotropic analytical algorithm, collapsed cone convolution, Monte Carlo , etc.). Pencil-beam algorithms that overestimate dose in heterogeneous tissue are generally not recommended.


    • Phantom-based QA on treatment plans.


    Dose Prescription






    • Dose and fractionation directed by adjacent normal tissue RT toxicity constraints with goal tumor BED10 > 100. Adaptive dosimetry for histology -, volume-, location-, and context-based lesions (primary vs. metastatic) are under investigation.


    • Current dose fractionation schema largely employs 1–5 fractions.


    • Peripheral Lung Tumors



      • Common accepted schemas: 25–34 Gy × 1 fraction, 18 Gy × 3 fractions, 12 Gy × 4 fractions, 10 Gy × 5 fractions.


    • Central Lung Tumors



      • We recommend: 10 Gy × 5 fractions (BED10 dose limited to reduce toxicity of central structures: large airways, heart , esophagus , and spinal cord ). See Fig. 7.1.

        A324326_1_En_7_Fig1_HTML.jpg


        Fig. 7.1.
        SBRT planning for a central early-stage NSCLC. Beam distribution shown on 3D anatomy reconstruction (left) and dose distribution for 50 Gy given in 5 fractions (right)


    • Dose typically prescribed 60–90 % IDL , with ≥95 % PTV coverage by prescription dose.


    • Composite planning should be employed in cases of regional lung re-irradiation with appropriate BED conversion for dose summation.


    Dose Limitations






    • See Table 7.2, assuming no prior regional radiotherapy (TG101, Benedict et al., 2010; RTOG 0618).


      Table 7.2
      Recommended dose constraints for SBRT lung lesion target planning




























































































      Structure

      Fractions

      Constraints

      Lung

      1

      V7 < 1500 cc

      3

      V11.6 < 1500 cc

      5

      V12.5 < 1500 cc

      Central airway

      1

      V10.5 < 4 cc, Dmax 20.2 Gy

      3

      V15 < 4 cc, Dmax 30 Gy

      5

      V16.5 < 4 cc, Dmax 40 Gy

      Chest wall

      1

      V22 < 1 cc, Dmax 30 Gy

      3

      V28.8 < 1 cc, Dmax 36.9 Gy

      5

      V35 < 1 cc, Dmax 43 Gy

      Heart

      1

      V16 < 15 cc, Dmax 22 Gy

      3

      V24 < 15 cc, Dmax 30 Gy

      5

      V32 < 15 cc, Dmax 38 Gy

      Esophagus

      1

      V11.9 < 5 cc, Dmax 15.4 Gy

      3

      V17.7 < 5 cc, Dmax 25.2 Gy

      5

      V19.5 < 5 cc, Dmax 35 Gy

      Brachial plexus

      1

      V14 < 3 cc, Dmax 17.5 Gy

      3

      V20.4 < 3 cc, Dmax 24 Gy

      5

      V 27 < 3 cc, Dmax 30.5 Gy

      Spinal cord

      1

      V10 < 0.35 cc, Dmax 14 Gy

      3

      V18 < 0.35 cc, Dmax 21.9 Gy

      5

      V23 < 0.35 cc, Dmax 30 Gy

      Skin

      1

      V23 < 10 cc, Dmax 26 Gy

      3

      V30 < 10 cc, Dmax 33 Gy

      5

      V36.5 < 10 cc, Dmax 39.5 Gy


    Dose Delivery






    • Dose delivered in consecutive daily or every other day fractions as per current NRG protocols.


    • Setup may be isocentric or non-isocentric depending upon SBRT delivery system.


    • Verification by kV XR or CBCT , aligned to visualized tumor or surrogate.


    • Intrafraction dose delivery adjustment by motion management and IGRT systems as discussed above.


    Toxicities and Management






    • Common acute toxicities (<6 weeks):



      • Fatigue



        • Generally early-onset and self-limiting.


        • Sustained fatigue may be related to cardiopulmonary dysfunction (CHF, CAD, COPD, etc.) and warrants further work up.


      • Cough/dyspnea



        • Low-grade cough common secondary to RT-related intrapulmonary inflammation. Antitussive pharmacotherapy for mild symptoms.


        • Severity of shortness of breath may be related to baseline lung function and associated comorbidities . For patients with moderate to severe symptoms or significant baseline comorbidities (COPD, ILD, CHF, etc.), recommend follow-up with pulmonology and/or cardiology.


      • Chest pain



        • May be related to regional pleuritis and/or pericarditis and is generally self-limited.


        • Analgesic pharmacotherapy recommended.


      • Pneumonitis



        • Associated with increased dose volume (V20 <10 %), smoking history (current/former), age, prior use of steroids , and comorbidity index on multiple studies.


        • Generally subacute onset (>2 weeks), associated with cough , dyspnea , hypoxia , and fever .


        • If symptomatic, treat with prednisone (1 mg/kg/d) or 60 mg/d and trimethoprim/sulfamethoxazole for PCP prophylaxis. Symptomatic relief may be rapid but slow steroid taper is critical for durable symptom resolution.


      • Esophagitis



        • Increased risk with treatment centrally located tumors, and is generally self-limited to several weeks after treatment.


        • Local or systemic analgesic pharmacotherapy (lidocaine , NSAIDs, opioids) ± proton pump inhibitor based on severity of symptoms.


      • Dermatitis



        • Chest wall entrance and exit doses can be reduced with increased numbers of beams to minimize radiation dermatitis.


        • Mild to moderate skin reaction treated with supportive care , including topical moisturizer s , analgesics , low-dose steroids , and antimicrobial salves.


    • Common late toxicities (>6 weeks):



      • Persistent cough /dyspnea



        • Recommend consultation with pulmonary medicine for consideration of long-term bronchodilator and anti-inflammatory therapy.


      • Radiation pneumonitis



        • Most commonly observed at ~6 weeks.


        • As above, recommend steroids with gradual taper for symptomatic patients


      • Brachial plexopathy



        • Apical lung tumors associated with greater risk of brachial plexus injury.


        • May present with neuropathic pain as seen in Lhermittes syndrome or with motor/sensory changes in the upper extremities.


        • MRI of brachialplexus and upper spine may be diagnostic and rule out tumor recurrence.


        • Limited treatment options include supportive care and occupational therapy .


      • Chest wall pain and rib fracture



        • More common in patients with peripheral lesions.


        • Supportive care indicated.


      • Radiation skin ulcer



        • For persistent non-healing skin lesions, consider hyperbaric oxygen therapy and tocopherol pharmacotherapy .


      • Esophageal stricture and tracheoesophageal fistula



        • Historically rare complication observed with treatment of mediastinal lymphadenopathy in locally advanced lung cancer .


        • Even less likely with SBRT , if airway and esophageal constraints maintained, with exception of re-irradiation setting.


      • Vasculopathy



        • Vascular erosion may lead to limited hemoptysis or massive hemorrhage and death (seen in re-irradiation setting of central lesions).


    Recommended Follow-Up






    • CT or PETCT every 3–4 months × 3 years, every 6 months × 2 years, every 12 months thereafter for routine follow-up.


    • Assessment with RECIST criteria of limited utility due to wide spectrum of evolving radiographic features following SBRT including diffuse and patchy GGO , consolidation, and/or fibrosis .


    • In general, radiographic changes include early inflammatory response (≤3 months) followed by resolution of FDG activity and late fibrosis (>6 months) in area of treated lesion which is often dynamic and may evolve over several years.


    • Persistent increase in size and density of treated tumor on interval CTs in the early post-treatment setting (<12 months) or new densities at later times (>12 months) should be considered suspicious for recurrence, with recommendation for increased frequency of CT, interval PET scan, and consideration of biopsy and/or surgical or radiotherapy salvage procedure.


    • Role of molecular imaging and circulating tumor markers is under investigation.


    Evidence



    Primary Lung Cancer






    • Evidence widely supports efficacy and safety of SBRT in early-stage NSCLC, with optimal patient selection criteria and dose schema emerging as studies mature.


    • CALGB 39904 (Bogart et al. 2010). Phase I dose-escalation study of 39 stage I (≤4 cm) NSCLC patients. 70 Gy in 29 decreased to 17 fractions. 92.3 % actuarial control, 82.1 % actuarial distant control. No late grade 3 or 4 toxicity.


    • Onishi et al. (2004). Initial report of retrospective Japanese multi-institutional series of 245 patients with stage I NSCLC treated with SBRT 18–75 Gy in 1–22 fractions with a median follow-up of 24 months. Grade ≥3 toxicity 2.4 %. LR at 3 years for BED  ≥ 100 vs. BED < 100 was 8.1 % vs. 26.4 %, p < 0.05 and OS was 88.4 % vs. 69.4 %, p < 0.05, establishing BED ≥ 100 as prescribing criterion.


    • Nordic Study Group (Baumann et al. 2009). Phase II study of SBRT in 57 patients with medically inoperable early-stage peripheral tumors (40 stage IA, 17 stage IB), treated with 45–66 Gy in 3 fractions. Estimated 3 year LC and OS were 88.4 % and 59.5 %, respectively. Distant metastatic rate 16 %. Risk of any failure increased in T2 vs. T1 tumors (41 % vs. 18 %, p = 0.027).


    • RTOG 0236 (Timmerman et al. 2010, Stanic et al. 2014). Phase II multicenter trial of 55 patients with medically inoperable early-stage (<5 cm) peripheral NSCLC (44 stage IA, 11 stage IB), treated with 54 Gy in 3 fractions SBRT . Three year primary tumor and involved lobe control was 98 %. Rate of distant failure 22 % at 3 years. OS 56 % at 3 years. Grade 3 and 4 toxicities were 12.7 % and 3.5 %, respectively. Poor baseline PFT not predictive of SBRT-related toxicity.


    • Timmerman et al. (2006), Farikis et al. (2009). Phase II study of SBRT at Indiana University for T1-2N0 medically inoperable NSCLC patients (n = 70), 60–66 Gy in 3 fractions. LC and OS at 3 years were 88.1 % and 42.7 %, respectively. Grade ≥3 toxicity rates of 10.4 % peripheral vs. 27.3 % central over a median follow-up of 50.2 months (p = 0.088).


    • JCOG 0403 (Nagata et al. 2012). Phase II trial of SBRT in early-stage NSCLC, stratified by medically operable/inoperable. In medically inoperable arm, 100 patients with stage IA disease received 48 Gy in 4 fractions. LC and OS at 3 years were 88 % and 59.9 %, respectively. For 64 medically operable patients, LC and OS at 3 years were 86.0 % and 76.0 %, respectively. Grade 3 pneumonitis 7 %, overall grade 4 toxicity 2 %.


    • RTOG 0618 (Timmerman et al. 2013). Phase II trial of 33 patients with medically operable early-stage peripheral NSCLC (<5 cm), treated with 60 Gy in 3 fractions. Completed accrual in 2010 with results presented at ASCO 2013 showing estimated 2 years primary tumor failure rate of 7.8 %, with a median follow-up of 25 months. Local failure , including ipsilateral lobe, was 19.2 %. PFS and OS at 2 years were estimated at 65.4 % and 84.4 %. Grade 3 toxicity was 16 %.


    • RTOG 0813. Phase I/II dose-escalation trial of medically inoperable centrally located (<2 cm of proximal bronchial tree) early-stage NSCLC (<5 cm). At the time of accrual completion, dose escalated to 60 Gy in 5 fractions. Closed to accrual at 120 patients in 2013. Results are pending.


    • RTOG 0915 (Videtic et al. 2013). Phase II randomized trial of 34 Gy in 1 fraction vs. 48 Gy in 4 fractions for medially inoperable early-stage peripheral NSCLC (<5 cm). Study completed accrual in 2011 with 94 patients. At 1 year, LC 97.1 % vs. 97.6 %; OS 85.4 % vs. 91.1 %, and PFS 78.0 % vs. 84.4 %. Adverse events were 9.8 % vs. 13.3 %. Based on the favorable toxicity, the 34 Gy in 1 fraction arm will be compared to the 54 Gy in 3 fractions arm of RTOG 0236 in a phase III setting.


    • Hoppe et al. (2008). Study of 50 stage I NSCLC patients treated with SBRT 60 Gy in 3 fractions or 44–48 Gy in 4 fractions with a median follow-up of 6 months. Skin toxicity was 38 % grade 1, 8 % grade 2, 4 % grade 3, and 2 % grade 4. Reduced number of beams, proximal distance to chest wall , and skin dose ≥50 % prescription dose were associated with increased risk of skin toxicity.


    • Mutter et al. (2012). Retrospective study of 128 early-stage NSCLC patients receiving SBRT 40–60 Gy in 3–5 fractions. With a median follow-up of 16 months, grade ≥2 chest wall toxicity was 39 % estimated at 2 years. On dosimetric analysis, grade ≥2 chest wall pain was associated with a V30Gy >70 cm3 within a 2 cm 2D-ipsilateral chest wall expansion.


    • ACOSOG Z4099/RTOG 1021. Phase III trial of SBRT vs. sublobar resection for high-risk operable, early-stage peripheral NSCLC (<3 cm). Terminated for poor accrual.


    • ROSEL Trial (VUMC, NCT00687986). Phase III trial of SBRT (60 Gy in 3 or 5 fractions) vs. surgery for stage IA peripheral NSCLC. Terminated for poor accrual.


    • STARS Trial (MDACC, NCT00840749). Phase III trial of SBRT 60 Gy in 3–4 fractions based on lesion location vs. surgery for stage I NSCLC. Terminated for poor accrual.


    • Grills et al. (2010). Retrospective comparison of 124 patients (95 % medically inoperable) T1-2 N0 NSCLC receiving wedge resection (n = 69) vs. SBRT (n = 58), 48–60 Gy in 4–5 fractions. No statistically significant differences in LRR (27 % wedge vs. 9 % SBRT, p > 0.16) or CSS (94 % wedge vs. 93 % SBRT, p = 0.53) noted at a median follow-up of 30 months. OS favored wedge resection (87 % wedge vs. 72 % SBRT, p = 0.01).


    • Crabtree et al. (2010). Retrospective comparison of stage I NSCLC patients receiving either surgery (n = 462) or SBRT (n = 76) for definitive care. Surgical candidates had fewer medical comorbidities . Thirty-five percent of surgical patients were upstaged on final pathology. OS 5 years 55 % surgery and OS 3 years 32 % with SBRT. In propensity matched analysis, no statistically significant difference in LC (88 % vs. 90 %) and OS (54 % vs. 38 %) at 3 years in surgery vs. SBRT groups.


    • SEER -Medicare analysis (Shirvani et al. 2012). Comparative outcomes of stage I NSCLC patients ≥60 years old, which demonstrated overall ranked outcomes as lobectomy > sublobar resection  > SBRT  > conventional EBRT  > observation. However, as treatment outcomes were likely influenced by patient selection and comorbidities , there was no difference in OS between SBRT and surgical modalities on propensity matched analysis, and EBRT remained inferior to SBRT.


    • Shah et al. (2013a, b). Cost-effectiveness comparison of surgical resection vs. SBRT for stage I NSCLC patients >65 years. For marginally operable patients, SBRT was most cost effective with a mean cost and quality-adjusted life expectancy of $42 k/8.0 years vs. lobectomy at $49 k/8.9 years. However, for completely operable candidates, lobectomy was found more cost effective, having an incremental cost-effectiveness ratio of $13 K/quality-adjusted life year .


    • Table 7.3 summarizes several multiple primarily retrospective series indicating local control rate s of 85–95 % at 3–5 years, and overall survival rates of 50–95 % at 3–5 years for early-stage NSCLC managed with SBRT . Some series include limited numbers of recurrent and metastatic patients.


      Table 7.3
      Selected studies of SBRT treated NSCLC patients










































































































      Study

      Patients

      Treatment

      LC/OS

      Notes

      Onishi et al. JRS-SBRTSG (IJROBP 2013)

      2226 patients stage I NSCLC

      32–70 Gy in 3–12 fractions, median BED 107 Gy (range 58–150 Gy)

      3 years LC/OS 85 %/72 %

      3 years LPFS 87 % T1, 72 % T2

      3 years OS 75 % BED  ≥ 100 Gy vs. 63 % BED < 100 Gy (p < 0.01)

      2.9 % grade ≥3

      Grills et al. Multi-institutional (JTO 2012)

      482 patients (505 tumors) T1-3N0 NSCLC,

      87 % medically inoperable

      20–64 Gy in 1–15 fractions, median 54 Gy in 3 fractions

      2 years LC/OS 94 %/60 %,

      LC 96 % BED  ≥ 105 vs. 85 % BED < 105 (p < 0.001)

      7 % grade ≥2 pneumonitis

      3 % rib fracture

      Shibamoto et al. Japan (IJROBP 2013, Cancer 2011)

      180 patients stage I NSCLC (120 medically inoperable, 60 operable)

      Volume-adapted

      44 Gy in 4 fractions <1.5 cm, 48 Gy in 4 fractions 1.5–3.0 cm, 52 Gy in 4 fractions >3.0 cm

      3 years LC/OS

      83 %/69 %, OS 74 % operable vs. 59 % inoperable, LC 86 % ≤3 cm vs. 73 % >3 cm

      5 years LC/OS

      82 %/68 %

      13 % grade ≥2 pneumonitis

      Uematsu et al. Japan (IJROBP 2001)

      50 patients T1-T2N0 NSCLC (21 medically inoperable, 29 operable)

      50–60 Gy in 5–10 fractions (18 patients received 40–60 Gy in 20–33 fractions prior to SBRT)

      3 years LC/OS

      94 %/66 %

      (86 % OS in medically operable subgroup)

      4 % rib fracture

      Stephans et al. Cleveland Clinic (JTO 2009)

      94 patients stage I NSCLC medically inoperable

      50 Gy in 5 fractions,

      60 Gy in 3 fractions

      1 year LC/OS

      97 %/83 % 50 Gy in 5 fractions, 100 %/77 % 60 Gy in 3 fractions

      2.2 % grade 2 pneumonitis

      10 % grade 1–2 chest wall toxicity

      Olsen et al. Wash U. (IJRBOP 2011)

      130 patients early-stage NSCLC

      Peripheral tumors 54 Gy in 3 fractions, central tumors 45–50 Gy in 5 fractions

      2 years LC

      91 % (54 Gy in 5 fractions)

      100 % (50 Gy in 5 fractions)

      50 % (45 Gy in 5 fractions)

      2 years OS

      85 % operable

      61 % inoperable

      16 % chest wall pain (grade 1–3)

      3 % grade 2 pneumonitis

      Modh et al. MSKCC (IJROBP 2013)

      107 central tumors (83 primary NSCLC, 10 recurrent, 14 metastatic)

      45–50 Gy in 4–5 Gx

      2 years LC/OS 72 %/56 %

      12 % grade ≥ 3

      Trakul et al. Stanford (IJROBP 2012a, b)

      111 patients (100 primary NSCLC, 11 metastatic)

      Volume-adapted iSABR (GTV  < 12 ml → BED  < 100, GTV ≥ 12 ml → BED ≥ 100)

      18–30 Gy in 1 fraction, 50–60 Gy in 4–5 fractions

      1 year LC/OS

      94 %/90 % in primary NSCLC subgroup,

      15 % DM rate,

      no difference in LC/OS by BED

      4.% grade 3

      Taremi et al. Canada (IJROBP 2012)

      108 patients, stage I NSCLC medically inoperable, 24 % w/o path diagnosis

      Peripheral tumors 48 Gy in 4 fractions or 54–60 in 3 fractions, central tumors 50–60 Gy in 8–10 fractions

      4 years LC/OS 92 %/30 %

      4 years distant DFS 83 %

      11 % grade 3

      Lagerwaard et al. VUMC (IJROBP 2008)

      206 patients T1-2N0

      Risk-adapted 60 Gy in 3 fractions (T1), 5 fractions (T1 near CW or T2), or 8 fractions (central)

      2 years LC/OS 83 %/64 %

      (LC 92 % T1, 71 % T2)

      2 years distant PFS 77 %

      3 % grade ≥3 pneumonitis

      2 % rib fracture

      1 % late CW pain

      Badiyan et al. Wash U. (RO 2013)

      120 patients (early-stage NSCLC and AIS)

      54 Gy in 3 fractions

      3 years LC/OS

      100 %/35 % AIS ,

      86 %/47 % NSCLC

      Not reported

      Bradley et al. Wash U. (IJROBP 2010)

      91 patients stage I/II NSCLC, medically inoperable

      Peripheral tumors 54 Gy in 3 fractions, central tumors 45 Gy in 5 fractions

      2 years LC/OS

      86 %/70 %

      3 % grade 2 pneumonitis

      4 % rib fracture

      1 % brachial plexopathy

      Palma et al. VUMC (IJROBP 2012)

      176 patients stage I NSCLC, severe COPD

      60 Gy in 3–5 fractions

      3 years LC/OS

      89 %/47 %

      3 % grade 3

      Chang et al. MDACC (RO 2012)

      130 patients stage I NSCLC

      50 Gy in 4 fractions

      2 years LC/OS

      98 %/78 %

      12 % grade 2–3 pneumonitis

      Griffioen et al. VUMC (RO 2013)

      62 patients with multiple synchronous primary early-stage NSCLC

      54–60 Gy in 3–8 fractions

      2 years LC/OS

      84 %/56 %

      4.8 % grade 3


    Role as Boost for Locally Advanced Lung Cancer




    Sep 16, 2016 | Posted by in GENERAL RADIOLOGY | Comments Off on Lung

    Full access? Get Clinical Tree

    Get Clinical Tree app for offline access