Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1704 word(s) 1704 2021-11-22 08:55:09 |
2 The format is correct + 411 word(s) 2115 2021-12-22 07:54:19 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Harada, D. Oligoprogression in Non-Small Cell Lung Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/17407 (accessed on 27 July 2024).
Harada D. Oligoprogression in Non-Small Cell Lung Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/17407. Accessed July 27, 2024.
Harada, Daijiro. "Oligoprogression in Non-Small Cell Lung Cancer" Encyclopedia, https://encyclopedia.pub/entry/17407 (accessed July 27, 2024).
Harada, D. (2021, December 21). Oligoprogression in Non-Small Cell Lung Cancer. In Encyclopedia. https://encyclopedia.pub/entry/17407
Harada, Daijiro. "Oligoprogression in Non-Small Cell Lung Cancer." Encyclopedia. Web. 21 December, 2021.
Oligoprogression in Non-Small Cell Lung Cancer
Edit

Oligometastatic disease (OMD) is cancer in an intermediate state between the localized early stage and the advanced metastatic stage. 

non-small cell lung cancer oligometastatic disease oligoprogressive disease

1. OPD in NSCLC with Driver Mutations

The progression pattern can be classified into three categories: (1) progression of CNS metastasis alone; (2) systemic progression with or without CNS; and (3) OPD [1]. The frequency of OPD varies depending on its definition and is estimated to be between 15–47% [1][2][3].
For the treatment of OPD in patients with NSCLC with driver mutation, the addition of LAT, such as stereotactic body radiotherapy (SBRT), while continuing the previous tyrosine kinase inhibitor (TKI) may be adopted. This is because SBRT is preferred over conventional radiotherapy (RT) or invasive surgical treatment as it requires less time for interruption of TKI. As for other LATs, surgery is an important treatment modality for resection of primary and metastatic lesions in OMD, and retrospective studies have revealed beneficial results of LATs in patients with OPD [4]. However, surgical invasion and acute complications may increase the risk of prolonged resumption time for subsequent TKI therapy beyond PD. A retrospective analysis reported that radiofrequency ablation (RFA) was applied to lung lesions in 2 of 46 OPD cases [5]. Furthermore, a retrospective analysis of cryotherapy for OPD showed survival benefits [6]. However, RFA and cryotherapy tend to have high incidences of complications, such as hemoptysis and pneumothorax/pleural effusion requiring drainage [7][8]. Thus, to standardize surgery, RFA and cryotherapy as LAT for OPD, issues such as the relatively high possibility of prolonging resumption time of subsequent therapy beyond PD and the bias in treatment outcomes among different physicians and facilities need to be considered and resolved [9].
In retrospective analysis, the addition of LAT to OPD, mainly during treatment with molecularly targeted agents, is expected to improve local control, suppress secondary metastasis, and prolong progression-free survival (PFS) and overall survival (OS) (Table 1) [10][2][5][11][12][13][14][15][16][17][18]. The median PFS in patients with NSCLC with driver mutation treated with LAT is generally expected to be 6 months or more, and there are reports of significantly longer overall survival than in patients who are not eligible for LAT.
Table 1. Outcomes of local ablative therapy for oligoprogressive disease.
Authors Driver Mutation Design n Induction Tx Intervention Mainte. Tx mPFS1
(m)
TTP from Intervention (m) Duration of Treatment (m) MST (m) MST from Intervention (m) Comment Ref
Gan et al. ALK fusion+ retro 14 crizo local ablative Tx (RT or OP) crizotinib 14 5.5 28 NA NA   [16]
13 not eligible for LAT 7.2 NA NA
Yu et al. EGFR m+ retro 18 gef or erlo local ablative Tx (RT or OP) gefitinib or erlotinib 19 10.0 NA NA 41.0 TTF from intervention: 22 m [10]
Weickhardt et al. ALK fusion+ (n = 38) EGFR m+ (n = 27) retro ALK rearrange (n = 15)
EGFR m+ (n = 10)
crizo
erlo
local Tx (RT or OP) crizotinib
erlotinib
9.0
12.0
6.2 NA NA NA   [2]
26 crizo or erlo Pts without LAT 12.8 NA
Qiu et al. EGFR m+ retro 46 EGFR-TKI local ablative Tx (RT or RFA) EGFR-TKI NA 7.0 NA 35.0 13.0   [5]
Chan et al. EGFR m+ retro 25 EGFR-TKI local ablative Tx (RT) same TKI NA 7 *1 NA NA 28.2 *2 *1: p = 0.0017. *2: HR:0.4 4[95%CI:0.21–0.92], p = 0.030 [11]
25 (matched cohort) chemotherapy 4.1 14.7
Rossi et al. EGFR m+ retro 30 EGFR-TKI local ablative Tx (RT) same TKI 13.8 6.7 *3 NA 37.3 *4 NA *3: HR: 0.54 [95%CI:0.24–1.18], p = 0.06. *4: p < 0.0001. *5: p = 0.0015 [12]
13 same TKI 12.3 3.1 20.1
88 2nd line treatment or BSC 8.9 NA 15.1
EGFR m+ without
intrinsic resistance to EGFR-TKI
retro 29 EGFR-TKI local ablative Tx (RT) same TKI NA     37.3 *5 NA
12 same TKI NA NA 20.1
64 2nd line treatment or BSC     21.9
Santarpia et al. EGFR m+ retro 36 EGFR-TKI high-dose radiation therapy EGFR-TKI 12.5 6.3   38.7     [13]
Schmid et al. EGFR T790M+ retro 13 osim local ablative Tx (RT or OP) osimertinib NA 6.7 19.6 28.0 NR *6 *6: p = 0.2 [18]
13 osimertinib 20.2
Weiss et al. EGFR m+ at least 6 m without PD P2 25 erlo SRT erlotinib NA 6.0 NA NA 29.0   [14]
Xu et al. EGFR m+ retro 206 EGFR-TKI local ablative Tx (RT or OP) EGFR-TKI 10.7   18.3 37.4     [15]
Friedes et al. NSCLC retro 253 chemo or TKI definitive RT same systemic Tx NA 7.9 NA NA NA TTF from intervention was 8.8 months. [17]
Kagawa et al. NSCLC retro 10 ICI local ablative Tx (RT or OP) ICI beyond PD (n = 6) 10.4 NA NA NA NR *7 *7: p = 0.456 [19]
28 no local therapy NA NR
BSC: best supportive care; chemo: chemotherapy; CNS: central nervous system; crizo: crizotinib; erlo: erlotinib; gef: gefitinib; ICI: immune checkpoint inhibitor; NA: not available; NR: not reached; NSCLC: non-small cell lung cancer; m+: mutation; m: month; mainte.: maintenance; MST: median survival time; OP: operation; osim: osimertinib; P2: phase II study; PD: progression disease; retro: retrospective study; RT: radiation therapy; TKI: tyrosine kinase inhibitor; TTF: time to treatment failure; TTP; time to progression; Tx: treatment; WT: wild type. The * superscript numbers indicate the test of comparison of each outcome in the corresponding clinical trials.

Although most retrospective studies suggested that a strategy of controlling TKI-resistant OPD by aggressive LAT is clinically beneficial, the following theoretical basis can be considered. LAT will aid in the prevention or treatment of symptoms and complications caused by growing tumors in the near future, may prevent secondary dissemination of TKI-resistant clones, and may allow the continuation of current TKI maintenance therapy [20]. For example, SBRT to asymptomatic brain metastasis in OPD may delay cranial symptoms and allow continuation of TKI therapy until systemic metastases, resulting in a survival benefit.

Based on the retrospective studies mentioned above, the U.S. National Comprehensive Cancer Network guidelines currently recommend a strategy of employing LAT for OPD [21]. A prospective study in this regard is also underway, and the results will clarify the evidence in the future [14]. HALT (NCT03256981) is a multicenter phase II/III study of 110 patients with advanced NSCLC with driver mutations who developed OPD after induction of TKI therapy [13]. The study design was to randomize patients to receive SBRT with continued TKI therapy or standard chemotherapy for up to three extracranial sites of OPD and to compare PFS as the primary endpoint.

2. OPD in NSCLC without Driver Mutations

There are few reports on the efficacy of continuing treatment beyond the PD after the addition of LAT for OPD in NSCLC without driver mutations. The ongoing STOP-NSCLC (NCT02756793) is a randomized, multicenter, phase II trial with a primary endpoint of PFS in patients with OPD (up to three lesions in a single organ, including the brain, for a total of up to five lesions) in NSCLC without driver mutations, comparing SBRT for OPD and continuation of current systemic therapy versus standard chemotherapy [22].
NCT03808662 is an open-label, randomized phase II study in patients with triple-negative breast cancer and stage IV NSCLC without EGFR/ALK mutation. The primary endpoint was the PFS, and the secondary endpoint was the OS. Characteristically, this trial is open to patients with medical conditions that preclude participation in other systemic therapies or drug trials. The purpose of this study was to determine whether receiving SBRT, when the subject’s metastatic tumor (up to five lesions, excluding the brain) is just beginning to grow, will prolong the time until the worsening of the disease. STOP-NSCLC, HALT, and NCT03808662 trials examined the efficacy and safety of SBRT of all progressions in patients with OPD after response to systemic chemotherapy, and the results of these trials may allow us to evaluate the efficacy and safety of a strategy of continuation of treatment beyond PD with the addition of LAT in OPD, with and without driver mutations.

3. OPD in NSCLC Patients Treated with Immune Checkpoint Inhibitor

Patients with EGFR and ALK mutations have been excluded from the clinical trials of combination therapy of immune checkpoint inhibitor (ICI) with cytotoxic chemotherapy due to their low efficacy, and the development of treatment has been mainly focused on patients with NSCLC without driver mutations. ICI with and without cytotoxic chemotherapy has also been reported to have a favorable prognosis in patients with OPD.
Rheinheimer et al. defined OPD as a localized treatment failure at one or two anatomic sites, with one to five progressive measurable lesions (according to RECIST 1.1). As for the characteristics of the development of OPD, when ICI monotherapy was compared between first- and second-line therapy, it was shown to be more frequent in first-line therapy (20 vs. 10%, p < 0.05), to occur at a later time (median 11 vs. 5 months, p < 0.01), to affect fewer organs (mean 1.1 vs. 1.5, p < 0.05), and to have fewer lesions (1.4 vs. 2.3, p < 0.05). The frequency by organ was 42% for mediastinal LNs, 39% for the brain, and 24% for the lung. Compared with multiple PD, OPD had a later onset (9 vs. 2 months, p < 0.001), a better prognosis (mean 26 vs. 13 months, p < 0.001), and more cases with high PD-L1 expression (p < 0.001) [23].
These retrospective analyses suggest that the incidence of OPD in patients with ICI is approximately 20%, the median time of onset is 7–9 months, and the prognosis in OPD is better than that in MPD. OPD sites were reported relatively frequently in the primary tumor, mediastinal LN, brain, and intrapulmonary and abdominal LNs and tended to be more frequent in patients with response to ICI, patients with PFS longer than 6 months, males, patients with EGFR wild-type mutation, and patients with a smoking history. However, the impact of the LAT on the OS is uncertain. In addition, these data are from retrospective observational studies and should be interpreted with caution, keeping in mind that there are various biases and differences in the definition of OPD (number of OPD, response to ICI, and minimum duration of treatment). Since the efficacy of ICI is higher in the first-line than in the later-line regimens, evidence from standard first-line treatment including ICI should be emphasized for OPD [24][25].
As for the mechanism by which radiation for OPD may enhance the clinical efficacy of ICI, as well as the evidence for the efficacy of LAT in patients with driver mutation, it has been shown that besides prolonging PFS2 by controlling OPD, radiation may also contribute to this strategy by modifying cell surface molecules to enhance the efficacy of immunotherapy, activating the innate immune system to produce an abscopal effect, and enhancing the efficacy of ICI by reducing the tumor volume [26][27][28].
Currently, there are several prospective clinical trials of LAT for OPD after ICI therapy in NSCLC and for continuation of ICI therapy beyond the PD. For example, the OLCSG 2001 [UMIN000041778] study is a single-arm phase II study that evaluates the efficacy and safety of adding localized radiation as LAT and continuing maintenance therapy beyond the PD in patients who develop OPD after initial immune-chemotherapy. The primary endpoint is 1-year survival rate, and the secondary endpoints are OS, PFS, safety, and post-treatment status. NCT04767009 is an open-label, multicenter, phase II study to evaluate the efficacy and safety of SBRT in patients with OPD, who were defined as those not requiring palliative irradiation and whose numbers of OPD are based on the opinion of the investigator, in NSCLC without driver mutation after an anti PD-1 inhibitor, followed by PD-1 inhibitor maintenance beyond the PD. The primary endpoints are safety and 1-year new lesion-free survival rate, and the secondary endpoints are PFS and OS. NCT04549428 is a multicenter, open-label, single-arm phase II study to evaluate the preliminary efficacy, safety, and tolerability of atezolizumab in combination with 8-Gy single-dose radiation therapy in patients who were diagnosed with stage IV NSCLC with OPD. The study considered patients who had up to four OPD lesions in up to three organs, excluding the brain and bone, with both anti-PD-1 agents and first-line cytotoxic chemotherapy, regardless of PD-L1 status. The primary endpoint is the ORR. NCT04517526 is a multicenter, phase II study to evaluate the efficacy and safety of platinum-based chemotherapy + bevacizumab + durvalumab and salvage SBRT for patients with stage IV NSCLC with EGFR mutations after the failure of first-line osimertinib. The primary endpoints are the PFS and OS.

4. Summary

Currently, ICI treatment has become the standard of care, and radiation therapy is expected to improve the efficacy of ICI by producing an abscopal effect and reducing tumor volume. This mechanism is expected to have a beneficial effect on LAT and subsequent continuation of the ICI beyond PD for OPD. Long-term survival in patients treated with ICI plus chemotherapy has been observed at a certain percentage. Hopefully, a multidisciplinary treatment strategy using ICI, cytotoxic agents, molecular-targeted agents, and radiation will reveal the possibility of long-term survival or cure in patients with metastatic NSCLC who do not respond adequately to ICI plus chemotherapy. There is also a need to establish clinically useful biomarkers to screen patients with rapid disease progression. Biomarkers in serum/plasma and tumor micro-environment may help detect the disease state [26].
In daily clinical practice, some patients meet the definition of OPD in clinical trials. They do not always receive LAT or subsequent continuation of the current therapy beyond PD. This is because established definitions and evidence of OPD are missing. It is important to conduct prospective comparative studies to verify the efficacy and safety, establish appropriate eligibility criteria, identify biomarkers, and determine the optimal dose and timing of LAT.

References

  1. Basler, L.; Kroeze, S.G.C.; Guckenberger, M. SBRT for oligoprogressive oncogene addicted NSCLC. Lung Cancer 2017, 106, 50–57.
  2. Weickhardt, A.J.; Scheier, B.; Burke, J.M.; Gan, G.; Lu, X.; Bunn, P.A.; Aisner, D.L.; Gaspar, L.E.; Kavanagh, B.D.; Doebele, R.C.; et al. Local Ablative Therapy of Oligoprogressive Disease Prolongs Disease Control by Tyrosine Kinase Inhibitors in Oncogene-Addicted Non–Small-Cell Lung Cancer. J. Thorac. Oncol. 2012, 7, 1807–1814.
  3. Al-Halabi, H.; Sayegh, K.; Digamurthy, S.R.; Niemierko, A.; Piotrowska, Z.; Willers, H.; Sequist, L.V. Pattern of Failure Analysis in Metastatic EGFR-Mutant Lung Cancer Treated with Tyrosine Kinase Inhibitors to Identify Candidates for Consolidation Stereotactic Body Radiation Therapy. J. Thorac. Oncol. 2015, 10, 1601–1607.
  4. Ashworth, A.B.; Senan, S.; Palma, D.A.; Riquet, M.; Ahn, Y.C.; Ricardi, U.; Congedo, M.T.; Gomez, D.R.; Wright, G.; Melloni, G.M.; et al. An Individual Patient Data Metaanalysis of Outcomes and Prognostic Factors After Treatment of Oligometastatic Non–Small-Cell Lung Cancer. Clin. Lung Cancer 2014, 15, 346–355.
  5. Qiu, B.; Liang, Y.; Li, Q.; Liu, G.; Wang, F.; Chen, Z.; Liu, M.; Zhao, M.; Liu, H. Local Therapy for Oligoprogressive Disease in Patients With Advanced Stage Non–small-cell Lung Cancer Harboring Epidermal Growth Factor Receptor Mutation. Clin. Lung Cancer 2017, 18, e369–e373.
  6. Niu, L.; Chen, J.; Yao, F.; Zhou, L.; Zhang, C.; Wen, W.; Bi, X.; Hu, Y.; Piao, X.; Jiang, F.; et al. Percutaneous cryoablation for stage IV lung cancer: A retrospective analysis. Cryobiology 2013, 67, 151–155.
  7. Hiraki, T.; Tajiri, N.; Mimura, H.; Yasui, K.; Gobara, H.; Mukai, T.; Hase, S.; Fujiwara, H.; Iguchi, T.; Sano, Y.; et al. Pneumothorax, Pleural Effusion, and Chest Tube Placement after Radiofrequency Ablation of Lung Tumors: Incidence and Risk Factors. Radiolog 2006, 241, 275–283.
  8. McDevitt, J.L.; Mouli, S.K.; Nemcek, A.A.; Lewandowski, R.J.; Salem, R.; Sato, K.T. Percutaneous Cryoablation for the Treatment of Primary and Metastatic Lung Tumors: Identification of Risk Factors for Recurrence and Major Complications. J. Vasc. Int. Radiol. 2016, 27, 1371–1379.
  9. Lam, A.; Yoshida, E.J.; Bui, K.; Katrivesis, J.; Fernando, D.; Nelson, K.; Abi-Jaoudeh, N. Demographic and facility volume related outcomes in radiofrequency ablation for early-stage hepatocellular carcinoma. HPB 2019, 21, 849–856.
  10. Yu, H.A.; Sima, C.S.; Huang, J.; Solomon, S.B.; Rimner, A.; Paik, P.; Pietanza, M.C.; Azzoli, C.G.; Rizvi, N.A.; Krug, L.M.; et al. Local Therapy with Continued EGFR Tyrosine Kinase Inhibitor Therapy as a Treatment Strategy in EGFR-Mutant Advanced Lung Cancers That Have Developed Acquired Resistance to EGFR Tyrosine Kinase Inhibitors. J. Thorac. Oncol. 2013, 8, 346–351.
  11. Chan, O.; Lee, V.; Mok, T.; Mo, F.; Chang, A.; Yeung, R. The Role of Radiotherapy in Epidermal Growth Factor Receptor Mutation-positive Patients with Oligoprogression: A Matched-cohort Analysis. Clin. Oncol. 2017, 29, 568–575.
  12. Rossi, S.; Finocchiaro, G.; Di Noia, V.; Bonomi, M.; Cerchiaro, E.; De Rose, F.; Franceschini, D.; Navarria, P.; Ceresoli, G.L.; Beretta, G.D.; et al. Survival outcome of tyrosine kinase inhibitors beyond progression in association to radiotherapy in oligoprogressive EGFR-mutant non-small-cell lung cancer. Futur. Oncol. 2019, 15, 3775–3782.
  13. Santarpia, M.; Altavilla, G.; Borsellino, N.; Girlando, A.; Mancuso, G.; Pergolizzi, S.; Piazza, D.; Pontoriero, A.; Valerio, M.R.; Gebbia, V. High-dose Radiotherapy for Oligo-progressive NSCLC Receiving EGFR Tyrosine Kinase Inhibitors: Real World Data. In Vivo 2020, 34, 2009–2014.
  14. Weiss, J.; Kavanagh, B.; Deal, A.; Villaruz, L.; Stevenson, J.; Camidge, R.; Borghaei, H.; West, J.; Kirpalani, P.; Morris, D.; et al. Phase II study of stereotactic radiosurgery for the treatment of patients with oligoprogression on erlotinib. Cancer Treat. Res. Commun. 2019, 19, 100126.
  15. Xu, Q.; Liu, H.; Meng, S.; Jiang, T.; Li, X.; Liang, S.; Ren, S.; Zhou, C. First-line continual EGFR-TKI plus local ablative therapy demonstrated survival benefit in EGFR-mutant NSCLC patients with oligoprogressive disease. J. Cancer 2019, 10, 522–529.
  16. Gan, G.N.; Weickhardt, A.J.; Scheier, B.; Doebele, R.C.; Gaspar, L.E.; Kavanagh, B.D.; Camidge, D.R. Stereotactic Radiation Therapy can Safely and Durably Control Sites of Extra-Central Nervous System Oligoprogressive Disease in Anaplastic Lymphoma Kinase-Positive Lung Cancer Patients Receiving Crizotinib. Int. J. Radiat. Oncol. 2014, 88, 892–898.
  17. Friedes, C.; Mai, N.; Fu, W.; Hu, C.; Hazell, S.Z.; Han, P.; McNutt, T.R.; Forde, P.M.; Redmond, K.J.; Voong, K.R.; et al. Isolated progression of metastatic lung cancer: Clinical outcomes associated with definitive radiotherapy. Cancer 2020, 126, 4572–4583.
  18. Schmid, S.; Klingbiel, D.; Aeppli, S.; Britschgi, C.; Gautschi, O.; Pless, M.; Rothschild, S.; Wannesson, L.; Janthur, W.; Foerbs, D.; et al. Patterns of progression on osimertinib in EGFR T790M positive NSCLC: A Swiss cohort study. Lung Cancer 2019, 130, 149–155.
  19. Kagawa, Y.; Furuta, H.; Uemura, T.; Watanabe, N.; Shimizu, J.; Horio, Y.; Kuroda, H.; Inaba, Y.; Kodaira, T.; Masago, K.; et al. Efficacy of local therapy for oligoprogressive disease after programmed cell death 1 blockade in advanced non-small cell lung cancer. Cancer Sci. 2020, 111, 4442–4452.
  20. Laurie, S.A.; Banerji, S.; Blais, N.; Brule, S.; Cheema, P.K.; Cheung, P.; Daaboul, N.; Hao, D.; Hirsh, V.; Juergens, R.; et al. Canadian Consensus: Oligoprogressive, Pseudoprogressive, and Oligometastatic Non-Small-Cell Lung Cancer. Curr. Oncol. 2019, 26, 81–93.
  21. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology (NCCN Guidelines). Non-Small Cell Lung Cancer, Version 3.2020. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 7 April 2020).
  22. Stereotactic Body Radiotherapy for the Treatment of OPD (HALT). Available online: https://clinicaltrials.gov/ct2/show/NCT03256981?term=HALT&draw=2&rank=7 (accessed on 18 September 2021).
  23. Rheinheimer, S.; Heussel, C.-P.; Mayer, P.; Gaissmaier, L.; Bozorgmehr, F.; Winter, H.; Herth, F.J.; Muley, T.; Liersch, S.; Bischoff, H.; et al. Oligoprogressive Non-Small-Cell Lung Cancer under Treatment with PD-(L)1 Inhibitors. Cancers 2020, 12, 1046.
  24. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833.
  25. Herbst, R.S.; Baas, P.; Kim, D.-W.; Felip, E.; Perez-Gracia, J.L.; Han, J.-Y.; Molina, J.; Kim, J.-H.; Arvis, C.D.; Ahn, M.-J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2016, 387, 1540–1550.
  26. Lin, A.J.; Roach, M.; Bradley, J.; Robinson, C. Combining stereotactic body radiation therapy with immunotherapy: Current data and future directions. Transl. Lung Cancer Res. 2018, 8, 107–115.
  27. Huang, A.C.; Postow, M.A.; Orlowski, R.J.; Mick, R.; Bengsch, B.; Manne, S.; Xu, W.; Harmon, S.; Giles, J.R.; Wenz, B.; et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017, 545, 60–65.
  28. Sharabi, A.B.; Lim, M.; DeWeese, T.L.; Drake, C.G. Radiation and checkpoint blockade immunotherapy: Radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015, 16, e498–e509.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 300
Revisions: 2 times (View History)
Update Date: 23 Dec 2021
1000/1000
Video Production Service