The second- and third-generations of ROS1 inhibitors have developed recently, but the ORR of crizotinib-resistant patients were not very satisfactory

The second- and third-generations of ROS1 inhibitors have developed recently, but the ORR of crizotinib-resistant patients were not very satisfactory. After 5 cycles of chemotherapy, CT scans revealed increased size of bilateral lobe nodules indicative of progressive disease (PD). Then the patient received treatment of crizotinib and his progression-free survival reached 3 months. Due to uncontrollable disease progression, the patient expired. Lessons: The genetic profile of NSCLC patients might be altered in various therapeutic processes. Thus, repeated genetic testing might be important at each progression. Moreover, immunotherapy might be a powerful weapon to overcome the resistance to Tyrosine kinase inhibitors (TKIs) in future. Keywords: crizotinib, immune checkpoint inhibitor, ROS1-fusion, TMB 1.?Introduction ROS1 fusions define a unique molecular subgroup of non-small cell lung cancer (NSCLC), accounting for approximately 2% of patients with NSCLC.[1] While most of these patients respond to crizotinib, resistance to this therapy eventually develops. Therefore, novel strategies are urgently needed. The second-generation ROS1 inhibitors have shown no obvious benefit in crizotinib-resistant NSCLC patients.[2,3] As the third-generation ROS1 inhibitor, lorlatinib showed a 50% objective response rate in ROS1-fusion NSCLC patients, irrespective of crizotinib resistance.[4] However, the role of immunotherapy in patients with crizotinib resistance has not been reported. Herein, we reported a significant elevation of tumor mutation burden (TMB) in a ROS1-fusion NSCLC patient who progressed on crizotinib. As TMB is a recognized predictive biomarker for response to immune checkpoint blockade in NSCLC,[5] we speculated that PD-1 or PD-L1 antibodies might be a fine selection for this patient. 2.?Case description A 44-year-old Chinese female presented with discontinuous dry cough and enlargement of left cervical lymph nodes in November 2017. A PET/CT scan revealed that: 1) a tumor in the left upper lobe with multiple small metastases in bilateral lung lobes 2) metastases in C6 and T9 vertebra; 3) metastasis in bilateral parietal lobes, left frontal lobe and cerebellum; and 4) metastases in bilateral hilar and mediastinal lymph nodes, bilateral cervical and supraclavicular lymph nodes. Then, ultrasound-guided left cervical lymph node biopsy revealed poorly differentiated lung adenocarcinoma, and immunohistochemistry (IHC) indicated the following phenotype: CK5/6(+), TTF-1(+), NapsinA(+), Ki-67(30%+), P63(spread+), P53(?), P40(?), and ALK-L(?) (Ventana method). Therefore, the staging of this patient was IVb stage (cT4N3M1c). EGFR mutations were not found in puncture biology of lymph node by genetic screening. Furthermore, a next-generation sequencing (NGS)-centered ctDNA genetic screening on blood also showed no mutations or rearrangement of EGFR, ALK, ROS1 and BRAF. Thus, the patient was treated with 4 cycles of pemetrexed, carboplatin and bevacizumab. CT and MRI scans performed 2 cycles of treatment indicated a partial remission (PR), and subsequent scans performed after 4 cycles of treatment showed a stable disease (SD). After 2 cycles of managed treatment of pemetrexed and bevacizumab, CT scans exposed increased size of the remaining top lobe nodules and growing pleural effusion indicative of progressive disease (PD) (Observe treatment timeline, Fig. ?Fig.11). Open in a separate window Number 1 Timeline of therapy in weeks including treatment process, genomic screening, and disease status. Bev?=?Bevacizumab, C?=?carboplatin, D?=?Doxtaxel, P?=?cisplatin, PD?=?progressive disease, Pem?=?pemetrexed, PR?=?partial response, SD?=?stable disease. Considering the resistance of first-line treatment, 1 cycle of docetaxel, cisplatin and bevacizumab was adopted on April 12, 2018. Meanwhile, the second NGS-based ctDNA genetic screening was performed on blood sample. Surprisingly, 22 novel gene mutation sites were revealed, including CD74-ROS1 fusion, CDKN2A, TP53 and so on (Fig. ?(Fig.2A).2A). Furthermore, TMB was also improved in this patient after chemotherapy (Fig. ?(Fig.2B).2B). Then, a CT scan exposed improved size of bilateral lobe nodules indicative of PD again, and the patient was switched to crizotinib. Crizotinib (250?mg bid, oral) therapy was initiated about April 30, 2018. After 4 weeks of crizotinib treatment, repeat CT imaging exposed dramatic decrease and shrinkage of bilateral lobe nodules and mediastinal lymph nodes. Thus, the patient achieved a confirmed partial remission (PR) after 4 weeks of crizotinib treatment. Open in a separate window Number 2 (A) Genomic alterations detected in samples from patient with ctDNA assays. (B) Alterations of TMB at each progression of treatment with ctDNA assays. Over the next 2 months, the patient continued on crizotinib treatment. Then, a CT scan exposed increased size of the remaining top lobe nodules and pleural effusion indicative of PD again. To determine the resistance mechanisms, the third.Recently, TMB has been recognized as a predictive biomarker for response to immune checkpoint blockade in many tumors. indicative of progressive disease (PD). Then the patient received treatment of crizotinib and his progression-free survival reached 3 months. Due to uncontrollable disease progression, the patient expired. Lessons: The genetic profile of NSCLC individuals might be modified in various restorative processes. Thus, repeated genetic testing might be important at each progression. Moreover, immunotherapy might be a powerful weapon to conquer the resistance to Tyrosine kinase inhibitors (TKIs) in long term. Keywords: crizotinib, immune checkpoint inhibitor, ROS1-fusion, TMB 1.?Intro ROS1 fusions define a unique molecular subgroup of non-small cell lung malignancy (NSCLC), accounting for approximately 2% of individuals with NSCLC.[1] While most of these individuals respond to crizotinib, resistance to this therapy eventually evolves. Therefore, novel strategies are urgently needed. The second-generation ROS1 inhibitors have shown no obvious benefit in crizotinib-resistant NSCLC individuals.[2,3] As the third-generation ROS1 inhibitor, lorlatinib showed a 50% objective response rate in ROS1-fusion NSCLC individuals, irrespective of crizotinib resistance.[4] However, the part of immunotherapy in individuals with crizotinib resistance has not been reported. Herein, we reported a significant elevation of tumor mutation burden (TMB) inside a ROS1-fusion NSCLC patient who progressed on crizotinib. As TMB is definitely a recognized predictive biomarker for response to immune checkpoint blockade in NSCLC,[5] we speculated that PD-1 or PD-L1 antibodies might be a fine selection for this patient. 2.?Case description A 44-year-old Chinese female presented with discontinuous dry cough and enlargement of left cervical lymph nodes in November 2017. A PET/CT scan revealed that: 1) a tumor in the left upper lobe with multiple small metastases in bilateral lung lobes 2) metastases in C6 and T9 vertebra; 3) metastasis in bilateral parietal lobes, left frontal lobe and cerebellum; and 4) metastases in bilateral hilar and mediastinal lymph nodes, bilateral cervical and supraclavicular lymph nodes. Then, ultrasound-guided left cervical lymph node biopsy revealed poorly differentiated lung adenocarcinoma, and immunohistochemistry (IHC) indicated the following phenotype: CK5/6(+), TTF-1(+), NapsinA(+), Ki-67(30%+), P63(scattered+), P53(?), P40(?), and ALK-L(?) (Ventana method). Thus, the staging of this patient was IVb stage (cT4N3M1c). EGFR mutations were not found in puncture biology of lymph node by genetic screening. Furthermore, a next-generation sequencing (NGS)-based ctDNA genetic screening on blood also showed no mutations or rearrangement of EGFR, ALK, ROS1 and BRAF. Thus, the patient was treated with 4 cycles of pemetrexed, carboplatin and bevacizumab. CT and MRI scans performed 2 cycles of treatment indicated a partial remission (PR), and subsequent scans performed after 4 cycles of treatment showed a stable disease (SD). After 2 cycles of managed treatment of pemetrexed and bevacizumab, CT scans revealed increased size of the left upper lobe nodules and emerging pleural effusion indicative of progressive disease (PD) (Observe treatment timeline, Fig. ?Fig.11). Open in a separate window Physique 1 Timeline of therapy in months including treatment process, genomic screening, and disease status. Bev?=?Bevacizumab, C?=?carboplatin, D?=?Doxtaxel, P?=?cisplatin, PD?=?progressive disease, Pem?=?pemetrexed, PR?=?partial response, SD?=?stable disease. Considering the resistance of first-line treatment, 1 cycle of docetaxel, cisplatin and bevacizumab was followed on April 12, 2018. In the mean time, the second NGS-based ctDNA genetic screening was performed on blood sample. Surprisingly, 22 novel gene mutation sites were revealed, including CD74-ROS1 fusion, CDKN2A, TP53 and so on (Fig. ?(Fig.2A).2A). Furthermore, TMB was also increased in this patient after chemotherapy (Fig. ?(Fig.2B).2B). Then, a CT scan revealed increased size of bilateral lobe nodules indicative of PD again, and the patient was switched to crizotinib. Crizotinib (250?mg bid, oral) therapy was initiated on April 30, 2018. After 4 weeks of crizotinib treatment, repeat CT imaging revealed dramatic decrease and shrinkage of bilateral lobe nodules and mediastinal lymph nodes. Thus, the patient achieved a confirmed partial remission (PR) after 4 weeks of crizotinib treatment. Open in a separate window Physique 2 (A) Genomic alterations detected in samples from patient with ctDNA assays. (B) Alterations of TMB at each progression of treatment with ctDNA assays. Over the next 2 months, the patient continued on crizotinib treatment. Then, a CT scan revealed increased size of the left upper lobe nodules and pleural effusion indicative of PD again. To determine the resistance mechanisms, the third NGS was performed on pleural fluid specimen and recognized an acquired ROS1 G2032R (4.1% MAF) mutation and KIT (11.5% MAF) mutation (Fig. ?(Fig.2A).2A). In particular, a significant elevation of TMB was observed in crizotinib-resistant sample compared with the pre-crizotinib one (21.0?Muts/Mb vs.Thus, the patient was treated with 4 cycles of pemetrexed, carboplatin and bevacizumab. The genetic profile of NSCLC patients might be altered in various therapeutic processes. Thus, repeated genetic screening might be important at each progression. Moreover, immunotherapy might be a powerful weapon to overcome the resistance to Tyrosine kinase inhibitors (TKIs) in future. Keywords: crizotinib, immune system checkpoint inhibitor, ROS1-fusion, TMB 1.?Intro ROS1 fusions define a distinctive molecular subgroup of non-small cell lung tumor (NSCLC), accounting for about 2% of individuals with NSCLC.[1] Some of these individuals react to crizotinib, level of resistance to the therapy eventually builds up. Therefore, book strategies are urgently required. The second-generation ROS1 inhibitors show no obvious advantage in crizotinib-resistant NSCLC individuals.[2,3] As the third-generation ROS1 inhibitor, lorlatinib showed a 50% goal response price in ROS1-fusion NSCLC individuals, regardless of crizotinib level of resistance.[4] However, the part of immunotherapy in individuals with crizotinib level of resistance is not reported. Herein, we reported a substantial elevation of tumor mutation burden (TMB) inside a ROS1-fusion NSCLC individual who advanced on crizotinib. As TMB can be an established predictive biomarker for response to immune system checkpoint blockade in NSCLC,[5] we speculated that PD-1 or PD-L1 antibodies may be an excellent selection because of this individual. 2.?Case explanation A 44-year-old Chinese language female offered discontinuous dry coughing and enhancement of still left cervical lymph nodes in November 2017. A Family pet/CT scan exposed that: 1) a tumor in the remaining top lobe with multiple little metastases in bilateral lung lobes 2) metastases in C6 and T9 vertebra; 3) metastasis in bilateral parietal lobes, remaining frontal lobe and cerebellum; and 4) metastases in bilateral hilar and mediastinal lymph nodes, bilateral cervical and supraclavicular lymph nodes. After that, ultrasound-guided remaining cervical lymph node biopsy exposed badly differentiated lung adenocarcinoma, and immunohistochemistry (IHC) indicated the next phenotype: CK5/6(+), TTF-1(+), NapsinA(+), Ki-67(30%+), P63(spread+), P53(?), P40(?), and ALK-L(?) (Ventana technique). Therefore, the staging of the individual was IVb stage (cT4N3M1c). EGFR mutations weren’t within puncture biology of lymph node by hereditary tests. Furthermore, a next-generation sequencing (NGS)-centered ctDNA genetic tests on bloodstream also demonstrated no mutations or rearrangement of EGFR, ALK, ROS1 and BRAF. Therefore, the individual was treated with 4 cycles of pemetrexed, carboplatin and bevacizumab. CT and MRI scans performed 2 cycles of treatment indicated a incomplete remission (PR), and following scans performed after 4 cycles of treatment demonstrated a well balanced disease (SD). After 2 cycles of taken care of treatment of pemetrexed and bevacizumab, CT scans exposed increased size from the remaining top lobe nodules and growing pleural effusion indicative of intensifying disease (PD) (Discover treatment timeline, Fig. ?Fig.11). Open up in another window Shape 1 Timeline of therapy in weeks including treatment procedure, genomic tests, and disease position. Bev?=?Bevacizumab, C?=?carboplatin, D?=?Doxtaxel, P?=?cisplatin, PD?=?intensifying disease, Pem?=?pemetrexed, PR?=?incomplete response, SD?=?steady disease. Taking into consideration the level of resistance of first-line treatment, 1 routine of docetaxel, cisplatin and bevacizumab was adopted on Apr 12, 2018. In the meantime, the next NGS-based ctDNA hereditary tests was performed on bloodstream test. Surprisingly, 22 book gene mutation sites had been revealed, including Compact disc74-ROS1 fusion, CDKN2A, TP53 etc (Fig. ?(Fig.2A).2A). Furthermore, TMB was also improved in this individual after chemotherapy (Fig. ?(Fig.2B).2B). After that, a CT scan exposed improved size of bilateral lobe nodules indicative of PD once again, and the individual was turned to crizotinib. Crizotinib CAL-130 Racemate (250?mg bet, dental) therapy was initiated about Apr 30, 2018. After four weeks of crizotinib treatment, do it again CT imaging exposed dramatic reduce and shrinkage of bilateral lobe nodules and mediastinal lymph nodes. Therefore, the patient accomplished a confirmed incomplete remission (PR) after four weeks of crizotinib treatment. Open up in another window Shape 2 (A) Genomic modifications detected in examples from individual with ctDNA assays. (B) Modifications of TMB at each development of treatment with ctDNA assays. More than another 2 months, the individual continuing on crizotinib treatment. After that, a CT scan exposed increased size from the remaining top lobe nodules and pleural effusion indicative of PD once again. To look for the level of resistance mechanisms, the 3rd NGS was performed on pleural liquid specimen and determined an obtained ROS1 G2032R (4.1% MAF) mutation and Package (11.5% MAF) mutation (Fig. ?(Fig.2A).2A). Specifically, a substantial elevation of TMB was seen in crizotinib-resistant test weighed against the pre-crizotinib one (21.0?Muts/Mb vs 5.8?Muts/Mb, Fig. ?Fig.2B).2B). Lately, TMB has.Sadly, the tumor created too fast to utilize the PD-1/PD-L1 antibodies. As the ROS1-fusion was discovered by next era sequencing, the individual received crizotinib treatment about three months. Results: After 5 cycles of chemotherapy, CT scans exposed improved size of bilateral lobe nodules indicative of intensifying CAL-130 Racemate disease (PD). Then your individual received treatment of crizotinib and his progression-free success reached three months. Because of uncontrollable disease development, the individual expired. Lessons: The hereditary profile of NSCLC individuals might be modified in various restorative processes. Therefore, repeated genetic tests might be essential at each development. Moreover, immunotherapy might be a powerful weapon to conquer the resistance to Tyrosine kinase inhibitors (TKIs) in long term. Keywords: crizotinib, immune checkpoint inhibitor, ROS1-fusion, TMB 1.?Intro ROS1 fusions define a unique molecular subgroup of non-small cell lung malignancy (NSCLC), accounting for approximately 2% of individuals with NSCLC.[1] While most of these individuals respond to crizotinib, resistance to this therapy eventually evolves. Therefore, novel strategies are urgently needed. The second-generation ROS1 inhibitors have shown no obvious benefit in crizotinib-resistant NSCLC individuals.[2,3] As the third-generation ROS1 inhibitor, lorlatinib showed a 50% objective response rate in ROS1-fusion NSCLC individuals, irrespective of crizotinib resistance.[4] However, the part of immunotherapy in individuals with crizotinib resistance has not been reported. Herein, we reported a significant elevation of tumor mutation burden (TMB) inside a ROS1-fusion NSCLC patient who progressed on crizotinib. As TMB is definitely a recognized predictive biomarker for response to immune checkpoint blockade in NSCLC,[5] we speculated that PD-1 or PD-L1 antibodies might be a fine selection for this patient. 2.?Case description A 44-year-old Chinese female presented with discontinuous dry cough and enlargement of left cervical lymph nodes in November 2017. A PET/CT scan exposed that: 1) a tumor in the remaining top lobe with multiple small metastases in bilateral lung lobes 2) metastases in C6 and T9 vertebra; 3) metastasis in bilateral parietal lobes, remaining frontal lobe and cerebellum; and 4) metastases in bilateral hilar and mediastinal lymph nodes, bilateral cervical and supraclavicular lymph nodes. Then, ultrasound-guided remaining cervical lymph node biopsy exposed poorly differentiated lung adenocarcinoma, and immunohistochemistry (IHC) indicated the following phenotype: CK5/6(+), TTF-1(+), NapsinA(+), Ki-67(30%+), P63(spread+), P53(?), P40(?), and ALK-L(?) (Ventana method). Therefore, the staging of this patient was IVb stage (cT4N3M1c). EGFR mutations were not found in puncture biology of lymph node by genetic screening. Furthermore, a next-generation sequencing (NGS)-centered ctDNA genetic screening on blood also showed no mutations or rearrangement of EGFR, ALK, ROS1 and BRAF. Therefore, the patient was treated with 4 cycles of pemetrexed, carboplatin and bevacizumab. CT and MRI scans performed 2 cycles of treatment indicated a partial remission (PR), and subsequent scans performed after 4 cycles of treatment showed a stable disease (SD). After 2 cycles of managed treatment of pemetrexed and bevacizumab, CT scans exposed increased size of the remaining top lobe nodules and growing pleural effusion indicative of progressive disease (PD) (Observe treatment timeline, Fig. ?Fig.11). Open in a separate window Number 1 Timeline of therapy in weeks including treatment process, genomic screening, and disease status. Bev?=?Bevacizumab, C?=?carboplatin, D?=?Doxtaxel, P?=?cisplatin, PD?=?progressive disease, Pem?=?pemetrexed, PR?=?partial response, SD?=?stable disease. Considering the resistance of first-line treatment, 1 cycle of docetaxel, cisplatin and bevacizumab was adopted on April 12, 2018. In the mean time, the second NGS-based ctDNA genetic screening was performed on blood sample. Surprisingly, 22 novel gene mutation sites were revealed, including CD74-ROS1 fusion, CDKN2A, TP53 and so on (Fig. ?(Fig.2A).2A). Furthermore, TMB was also improved in this patient after chemotherapy (Fig. ?(Fig.2B).2B). Then, a CT scan exposed improved size of bilateral lobe nodules indicative of PD again, and the patient was switched to crizotinib. Crizotinib (250?mg bid, oral) therapy was initiated about Apr 30, 2018. After four weeks of crizotinib treatment, do it again CT imaging uncovered dramatic reduce and shrinkage of bilateral lobe nodules and mediastinal lymph nodes. Hence, the patient attained a confirmed.Comprehensive studies have explored the fundamental mechanisms of crizotinib Vegfb resistance, including ROS1 mutations,[8,9] KIT mutation,[10] BRAF CDKN2A and mutation[11] mutation.[12] Within this report, a ROS was found by us G2032R mutation-induced level of resistance, and PFS was just 3.0 months. by following generation sequencing, the individual received crizotinib treatment about three months. Final results: After 5 cycles of chemotherapy, CT scans uncovered elevated size of bilateral lobe nodules indicative of intensifying disease (PD). Then your individual received treatment of crizotinib and his progression-free success reached three months. Because of uncontrollable disease development, the individual expired. Lessons: The hereditary profile of NSCLC sufferers might be changed in various healing processes. Hence, repeated genetic examining might be essential at each development. Moreover, immunotherapy may be a powerful tool to get over the level of resistance to Tyrosine kinase inhibitors (TKIs) in upcoming. Keywords: crizotinib, immune system checkpoint inhibitor, ROS1-fusion, TMB 1.?Launch ROS1 fusions define a distinctive molecular subgroup of non-small cell lung cancers (NSCLC), accounting for about 2% of sufferers with NSCLC.[1] Some of these sufferers react to crizotinib, level of resistance to the therapy eventually grows. Therefore, book strategies are urgently required. The second-generation ROS1 inhibitors show no obvious advantage in crizotinib-resistant NSCLC sufferers.[2,3] As the third-generation ROS1 inhibitor, lorlatinib showed a 50% goal response price in ROS1-fusion NSCLC sufferers, regardless of crizotinib level of resistance.[4] However, the function of immunotherapy in sufferers with crizotinib level of resistance is not reported. Herein, we reported a substantial elevation of tumor mutation burden (TMB) within a ROS1-fusion NSCLC individual who advanced on crizotinib. As TMB is normally an established predictive biomarker for response to immune system checkpoint blockade in NSCLC,[5] we speculated that PD-1 or PD-L1 antibodies may be an excellent selection because of this individual. 2.?Case explanation A 44-year-old Chinese language female offered discontinuous dry coughing and enhancement of still left cervical lymph nodes in November 2017. A Family pet/CT scan uncovered that: 1) a tumor in the still left higher lobe with multiple little metastases in bilateral lung lobes 2) metastases in C6 and T9 vertebra; 3) metastasis in bilateral parietal lobes, still left frontal lobe and cerebellum; and 4) metastases in bilateral hilar and mediastinal lymph nodes, bilateral cervical and supraclavicular lymph nodes. After that, ultrasound-guided still left cervical lymph node biopsy uncovered badly differentiated lung adenocarcinoma, and immunohistochemistry (IHC) indicated the next phenotype: CK5/6(+), TTF-1(+), NapsinA(+), Ki-67(30%+), P63(dispersed+), P53(?), P40(?), and ALK-L(?) (Ventana technique). Hence, the staging of the individual was IVb stage (cT4N3M1c). EGFR mutations weren’t within puncture biology of lymph node by hereditary examining. Furthermore, a next-generation sequencing (NGS)-structured ctDNA genetic examining on bloodstream also demonstrated no mutations or rearrangement of EGFR, ALK, ROS1 and BRAF. Hence, the individual was treated with 4 cycles of pemetrexed, carboplatin and bevacizumab. CT and MRI scans performed 2 cycles of treatment indicated a incomplete remission (PR), and following scans performed after 4 cycles of treatment demonstrated a well balanced disease (SD). After 2 cycles of preserved treatment of pemetrexed and bevacizumab, CT scans uncovered increased size from the still left higher lobe nodules and rising pleural effusion indicative of intensifying disease (PD) (Find treatment timeline, Fig. ?Fig.11). Open up in another window Amount 1 Timeline of therapy in a few months including treatment procedure, genomic examining, and disease position. Bev?=?Bevacizumab, C?=?carboplatin, D?=?Doxtaxel, P?=?cisplatin, PD?=?intensifying disease, Pem?=?pemetrexed, PR?=?incomplete response, SD?=?steady disease. Taking into consideration the level of resistance of first-line treatment, 1 routine of docetaxel, cisplatin and bevacizumab was CAL-130 Racemate implemented on Apr 12, 2018. On the other hand, the next NGS-based ctDNA hereditary examining was performed on bloodstream test. Surprisingly, 22 book gene mutation sites had been revealed, including Compact disc74-ROS1 fusion, CDKN2A, TP53 etc (Fig. ?(Fig.2A).2A). Furthermore, TMB was also elevated in this individual after chemotherapy (Fig. ?(Fig.2B).2B). After that, a CT scan uncovered elevated size of bilateral lobe nodules indicative of PD once again, and the individual was turned to crizotinib. Crizotinib (250?mg bet, dental) therapy was initiated in Apr 30, 2018. After four weeks of crizotinib treatment, do it again CT imaging uncovered dramatic reduce and shrinkage of bilateral lobe nodules and mediastinal lymph nodes. Hence, the patient attained a confirmed incomplete remission (PR) after four weeks of crizotinib treatment. Open up in another window Amount 2 (A) Genomic modifications detected in samples from patient with ctDNA assays. (B) Alterations of TMB at each progression of treatment with ctDNA assays. Over the next 2 months, the patient continued on crizotinib treatment. Then, a CT.