PSC is a rare type of NSCLC that contains sarcomatoid cells or sarcomatoid differentiation. According to the 2015 World Health Organization (WHO) classification, PSC can be divided into five subgroups: pleomorphic carcinoma (PC), spindle cell carcinoma (SCC), giant cell carcinoma (GCC) carcinosarcoma, and pulmonary blastoma[6]. PSC patients are mostly middle-aged or elderly men who smoke, with no specific clinical symptoms. Most patients develop metastases at early stages and cannot be operated on, which leads to poor prognosis. In an analysis of clinical characteristics of 38 patients with PSC, median survival was 21 months, and 1-year, 3-year, and 5-year survival rates were 68.4%, 31.6%, and 18.4%, respectively[7]. The 5-year survival rate was extremely low compared to other types of NSCLC.
Advanced PSC patients are highly resistant to first-line chemotherapy. A retrospective study showed that median PFS and median overall survival (OS) were 2.7 and 4.3 months respectively for PSC patients treated with first-line chemotherapeutic drugs[8]. How to improve the prognosis of PSC patients is a difficult problem. At present, a variety of gene mutations have been found in PSC patients, of which EGFR, TP53, KRAS, ALK and MET are the most common. Lococo et al. analyzed mutations in 49 PSC patients and found that 39 (80%) patients had at least one mutation, and that patients with PSC mutations had a shorter survival compared to those without mutations[9]. Another analysis showed that of 33 PSC patients, 72% had at least one genetic mutation, 58% had TP53 mutations, and 30% had KRAS mutations[10]. Such findings can be used as a reference for targeted therapy. It has been reported that afatinib combined with crizotinib could lead to partial remission in PSC patients[11]. Unfortunately, retrospective studies have shown that the majority of Chinese population cannot benefit from EGFR-TKI drugs due to low EGFR mutation rates. There are only few reports of the use of targeted drugs for other targets in PSC patients; therefore, the efficacy of targeted therapy in PSC patients is still largely unknown.
The rapid development of immunotherapy in recent years has changed the direction of tumor therapy. A retrospective study suggested that PD-L1 expression in PSC patients was higher than that in NSCLC patients, with a significant immune infiltration [12]. The expression of PD-L1 in tumor tissues has become a common and preferable biomarker for predicting the efficacy of immunotherapy. The high expression of PD-L1 provides a biological basis for immunotherapy. In a report of 5 patients with advanced PSC, the response rate after immunotherapy was 80%, the median OS was between 14 and 33 months, and one patient was fully responsive[13]. In multiple case reports, first-line immunotherapy has resulted in a variety of benefits for patients with PSC(Table 1). Registration trials of immunotherapy for PSC patients are also listed in this article (Table 2). However, in our case, PD-L1 expression was negative (TPS < 1%). In a retrospective study of 37 PSC patients treated with nivolumab as second or third line, the ORR and OS were 0% and 1.84 months in patients with negative PD-L1 expression, respectively[5]. Considering the high resistance of PSC patients to first-line chemotherapy, the absence of EGFR, ALK and other genes’ mutations, the low expression of PD-L1, and the characteristics of MSS, we believe that the application and benefits of PD-1 inhibitor monotherapy would be limited in this case. Therefore, we adopted the treatment strategy of combination therapy to fit the specific needs of this patient.
Table 1
Case reports of first-line treatment for pulmonary sarcomatoid carcinoma.
First author(year)
|
Number of
patients
|
Pathological
diagnosis(staging)
|
PD-L1
expression
|
Treatment regimen
|
PFS, months
|
OS, months
|
Yoko Matsumoto (2017)[24]
|
1
|
PPC, ⅣB
|
PD-L1 (+), >50%
|
pembrolizumab
|
2.5+
|
NA
|
Tozuka
(2018)[25]
|
1
|
PPC, IV
|
PD-L1 (+), >50%
|
pembrolizumab
|
5
|
NA
|
Xiaofeng Li (2018)[26]
|
1
|
PSC, IV
|
NA
|
apatinib
|
14+
|
NA
|
Hirokazu Tokuyasu
(2019)[27]
|
1
|
PPC, IV
|
PD-L1 (+), 100%
|
pembrolizumab
|
17+
|
NA
|
Vineeth Sukrithan
(2019)[13]
|
5
|
PSC
|
PD-L1 (+), >75%
|
pembrolizumab
|
11+-29+
|
14+-33+
|
Federica D’Antonio
(2019)[28]
|
1
|
PSC, IV
|
PD-L1 (+), >50%
|
pembrolizumab
|
3
|
NA
|
Emanuela Cimpeanu
(2020)[29]
|
1
|
PSC, IIIA
|
PD-L1 (+), >50%
|
pembrolizumab
|
14+
|
NA
|
Feng-Wei Kong (2020)[30]
|
3
|
PSC, III-IV
|
PD-L1 (+)
|
nab-paclitaxel+ carboplatin+ apatinib
|
6+,6+,7
|
NA
|
Fengwei Kong (2020)[31]
|
1
|
PSC, IIIB
|
PD-L1 (+)
|
camrelizumab +doxorubicin
|
20+
|
NA
|
Hirokazu Taniguchi (2021)[32]
|
1
|
PSC, ⅣB
|
PD-L1 (+), 1%
|
Pembrolizumab+ carboplatin+
pemetrexed
|
3
|
NA
|
OS=overall survival, PFS=progression-free survival, PSC=pulmonary sarcomatoid carcinoma, PPC=pulmonary pleomorphic carcinoma |
Table 2
The registered trials of immunotherapy therapy for PSC patients
Identifier
|
Year
|
Study design
|
Regimen
|
Estimated
enrollment
|
Treatment
lines
|
Primary
endpoint
|
Status
|
Country
|
NCT02834013
|
2016
|
Single-arm
|
Nivolumab and Ipilimumab or nivolumab
monotherapy
|
818
|
2nd line
and
beyond
|
ORR
|
Recruiting
|
America
|
NCT03022500
|
2017
|
Single-arm,
phase II
study
|
durvalumab+ tremelimumab
|
18
|
1st line
and
beyond
|
Response rate
|
Active, not recruiting
|
South Korea
|
NCT04224337
|
2020
|
Single-arm,
phase II
study
|
Durvalumab+ doxorubicin+ ifosfamide
|
34
|
1st line
and
beyond
|
Response rate
|
Recruiting
|
South Korea
|
ChiCTR2000031478
|
2020
|
Single-arm,
phase II
study
|
Camrelizumab +albumin-paclitaxel and
carboplatin
|
15
|
2nd line
|
ORR, DCR,
PFS
|
Recruiting
|
China
|
NCT04725448
|
2021
|
Single-arm,
phase II
study
|
Toripalimab +Bevacizumab,Nab-paclitaxel +Carboplatin
|
27
|
1st line
|
PFS
|
Recruiting
|
China
|
ORR=objective response rate, DCR=disease control rate, PFS=progression-free survival. |
In general, tumor growth and metastasis are dependent on neovascularization, and hypoxic-driven vascular endothelial growth factor (VEGF) is a major regulator of angiogenesis. The response to immunotherapy is associated with the immune invasion of tumor microenvironment (TME). However, VEGF blocks T cells infiltration by down-regulating lymphocytes, interferes with T cells functions [14, 15], promotes myeloid-derived suppressor cell (MDSCs), induces proliferation and differentiation of regulatory T cells (Tregs), and inhibits maturation of dendritic cells (DCs) precursors, which lead to immunosuppression of the TME[15, 16]. Anti-angiogenic drugs can reverse the immunosuppressive effect caused by VEGF, and also normalize the tumor vascular system and promote the delivery of T cells and other immune effector molecules. On the other hand, ICIs can normalize the tumor vascular system and increase the infiltration and killing function of effector T cells by activating them[17, 18]. In addition, vascular normalization can improve the efficiency of drug delivery. This combination therapy form is being explored in multiple solid tumors.
Impower150 is the first phase III study of the combination of immunotherapy and anti-angiogenic agents in NSCLC. In all patients, the combination group (atezolizumab + bevacizumab + carboplatin + paclitaxel, ABCP) showed significant improvement in PFS and OS, with median PFS of 8.4 months and median OS of 19.5 months compared to 6.8 months, and 14.7 months in (bevacizumab + carboplatin + paclitaxel, BCP) group, respectively[19]. Based on the research results of IMpower150, ABCP four-drug combination has become the first-line treatment recommendation for non-squamous NSCLC in the National Comprehensive Cancer Network guidelines of the United States. In the JVDF clinical trial, ramucirumab combined with pembrolizumab treated NSCLC with a median PFS of 9.7 months [20]. Meanwhile, at the 2019 World Lung Cancer Congress, Professor Baohui Han reported the efficacy of sintilimab combined with anlotinib as first-line treatment of advanced NSCLC with negative driver genes, with an objective response rate (ORR) of 72.7%, a disease control rate (DCR) of 100%, and a median PFS of 15 months [21]. This treatment regimen has shown good results in NSCLC patients, but its application in PSC has not been reported, and, therefore, improving the prognosis of PSC is still a challenge. At this year's ASCO meeting, the IMbrave150 study published the results of first-line treatment of patients with hepatocellular carcinoma (HCC) with atezolizumab combined with bevacizumab, the median OS was 19.2 months for all patients and 24.0 months for the Chinese subpopulation[22]. In the REGONIVO study, the combination of regorafenib and nivolumab overcame the disadvantage of immunotherapy in MSS type tumors [23]. Previously, MSS-type tumors have been at a disadvantage to benefit from immunotherapy. The successful finding of REGONIVO study is believed to be that regorafenib, by blocking multiple targets, can influence immune-related mechanisms and reverse anti-tumor immune activities. Similarly, anlotinib, as a novel multi-target antiangiogenic agent, has long been approved as a third-line treatment for NSCLC. Despite the low expression of PD-L1 and the characteristics of MSS, our patient still benefited from this combination therapy. The patient’s PFS reached 24 months and the esophageal lesion was improved, which far exceeded the benefits previously reported in PSC patients. Our findings present this combination therapy as a potential treatment for advanced PSC patients with negative driver-gene mutations. However, since this is the first investigation of this combination therapy in PSC, and due the inclusion of only one case in this report, future studies with larger sample sizes are needed to investigate the feasibility and efficacy of this scheme.
Combining immunotherapy with antiangiogenic agents as anti-tumor therapeutic strategy has enhanced patients’ survival in a variety of solid tumors. However, a further investigation of best drug combinations, doses, and treatment plans are still encouraged. A further consideration of improvements to increase efficacy, yet reduce toxicity and tolerance in patients is also needed in future studies. In addition, no biomarker has been found that can accurately predict the efficacy of such therapeutic combination. Such problems should be addressed and investigated to improve the application of the combination of immunotherapy and anti-angiogenic drugs.