ArticlePDF Available

Leptomeningeal metastasis from non-small cell lung cancer – a post-hoc analysis from four randomised clinical trials

Authors:

Abstract and Figures

Background: Leptomeningeal metastasis (LMM) from non-small cell lung cancer (NSCLC) is often an underdiagnosed entity, has a dismal prognosis and has very limited data from low- and middle-income countries. Methods: A single-centre study, which included 1148 adult patients diagnosed as NSCLC, with Eastern Oncology Cooperative Group performance status 0-2, as identified from data of four prospective randomised controlled trials. Two trials included patients who had epidermal growth factor sensitive mutations (CTRI/2015/08/006113 and CTRI/2016/08/007149) and the other two included squamous cell carcinoma (CTRI/2013/02/003422) and adenocarcinoma patients (CTRI/2014/08/00484). The key objectives were to estimate the incidence, risk factors, time to development and outcomes for LMM. Results: Out of 1148 patients, 36 patients (0.031%; 95%CI: 0.022-0.043) developed leptomeningeal metastasis. In these patients, median time to development of LM was 14.92 months (interquartile range: 7.7-21.84). Among the tested factors, the presence of brain metastasis was the only statistically significant risk factor associated with the development of LMM (p-value = 0.035). The median overall survival (OS) after the development of LM was 61 days (95%CI: 38.95-83.05). The median OS in driver mutated patients was 66 days (95% CI: 14.74-117.26) versus 51 days (95% CI: 14.5-87.5) (p-value = 0.201) in non-driver mutated patients. Only 6 (19.4%) out of 31 epidermal growth factor receptor-mutated patients received osimertinib. Patients treated with osimertinib had a median OS of 245 days (95% CI: 215.48-274.52) versus 52 days (95% CI: 22.62-81.38) for those without (p-value = 0.327). Conclusion: The incidence of LMM is low in the Indian population. In our study, there was no single factor which impacted survival in patients who developed LMM. This suggests that the overall prognosis is poor in patients with LMM where access to newer therapeutic modalities is limited.
Content may be subject to copyright.
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 1
Research
Leptomeningeal metastasis from non-small cell lung cancer—
a post-hoc analysis from four randomised clinical trials
Vijay Patil1†, Vanita Noronha1†, Dilip Harindran Vallathol1†, Nandini Menon1, Abhishek Mahajan2, Amit Janu2, Nilendu Purandare3 and
Kumar Prabhash1
1Department of Medical Oncology, Tata Memorial Centre, HBNI, Mumbai 400012, India
2Department of Radiology, Tata Memorial Centre, HBNI, Mumbai 400012, India
3Department of Nuclear Medicine, Tata Memorial Centre, HBNI, Mumbai 400012, India
Both contributed equally.
Correspondence to: Kumar Prabhash
Email: kumarprabhashtmh@gmail.com
ecancer 2022, 16:1414
https://doi.org/10.3332/ecancer.2022.1414
Published: 16/06/2022
Received: 21/08/2021
Publication costs for this article were supported by
ecancer (UK Charity number 1176307).
Copyright: © the authors; licensee
ecancermedicalscience. This is an Open Access
article distributed under the terms of the
Creative Commons Attribution License (http://
creativecommons.org/licenses/by/4.0), which
permits unrestricted use, distribution, and
reproduction in any medium, provided the original
work is properly cited.
Abstract
Background: Leptomeningeal metastasis (LMM) from non-small cell lung cancer (NSCLC)
is often an underdiagnosed entity, has a dismal prognosis and has very limited data from
low- and middle-income countries.
Methods: A single-centre study, which included 1148 adult patients diagnosed as
NSCLC, with Eastern Oncology Cooperative Group performance status 0–2, as identi-
fied from data of four prospective randomised controlled trials. Two trials included
patients who had epidermal growth factor sensitive mutations (CTRI/2015/08/006113
and CTRI/2016/08/007149) and the other two included squamous cell carcinoma
(CTRI/2013/02/003422) and adenocarcinoma patients (CTRI/2014/08/00484). The
key objectives were to estimate the incidence, risk factors, time to development and
outcomes for LMM.
Results: Out of 1148 patients, 36 patients (0.031%; 95%CI: 0.022–0.043) developed
leptomeningeal metastasis. In these patients, median time to development of LM was
14.92 months (interquartile range: 7.7–21.84). Among the tested factors, the presence
of brain metastasis was the only statistically significant risk factor associated with the
development of LMM (p-value = 0.035). The median overall survival (OS) after the devel-
opment of LM was 61 days (95%CI: 38.95–83.05). The median OS in driver mutated
patients was 66 days (95% CI: 14.74–117.26) versus 51 days (95% CI: 14.5–87.5)
(p-value = 0.201) in non-driver mutated patients. Only 6 (19.4%) out of 31 epidermal
growth factor receptor-mutated patients received osimertinib. Patients treated with
osimertinib had a median OS of 245 days (95% CI: 215.48–274.52) versus 52 days
(95% CI: 22.62–81.38) for those without (p-value = 0.327).
Conclusion: The incidence of LMM is low in the Indian population. In our study, there
was no single factor which impacted survival in patients who developed LMM. This sug-
gests that the overall prognosis is poor in patients with LMM where access to newer
therapeutic modalities is limited.
Keywords: leptomeningeal metastasis, non-small cell lung cancer, incidence, survival
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 2
Introducon
Leptomeningeal metastasis (LMM) is a rare and underdiagnosed entity in extracranial solid tumours [1, 2]. It often occurs in the setting of
a progression of systemic disease. The estimated incidence of LMM in solid tumours (excluding central nervous system tumours) is around
5%–10% [3]. The treatment of LMM has also evolved, but the prognosis of LMM remains dismal when compared to other sites of metas-
tasis [4]. The two commonest extracranial solid tumour sites leading to LMM are breast carcinoma and non-small cell lung cancer (NSCLC)
[3]. The improvement in diagnostic modalities and newer modalities of treatment of malignancies has led to an increase in the diagnosis of
LMM [5, 6].
The occurrence of LMM is particularly increased in patients with driver mutations as a result of their improved outcomes with targeted
therapies and better extracranial disease control [5, 6]. Previously reported studies by Yang et al [6] and Liao et al [7] in a similar setting
showed higher rates of LMM among epidermal growth factor receptor (EGFR)-mutated patients when compared to mutation negative
patients. Yang et al [6] showed a 9.4% incidence of LMM among EGFR-mutated patients when compared to 1.7% in negative ones; Liao et
al [7] reported 212 patients (3.8%) with LMM in NSCLC of which 75 patients harboured EGFR mutations. The mechanisms responsible for
the higher frequency of LMM in patients with driver mutations may be multifactorial. Longer survival and insufficient penetration of tyrosine
kinase inhibitors (TKIs) into the CSF are likely to be the main explanations. With the application of TKIs, overall survival (OS) of metastatic
NSCLC with driver mutations was improved in comparison to the OS of patients with wild-type. Moreover, the ratio of the CSF to plasma
concentration of TKIs is low on account of incomplete penetration of the blood–brain barrier [8]. Hence, insufficient levels of TKIs, which
result in continuing lower stimulation of tumour cells, may favour resistance and pave the way for LMM. Even though these are postulated,
the exact mechanism has not yet been elucidated. Globally, the improvement in treatment of malignancies, with the entry of novel therapies
and higher generation of targeted agents with better central nervous system penetration, has improved the median survival in LMM from
1–3 months to 3–11 months [5, 6]. However, access to these novel agents is limited in low- and middle-income countries (LMICs). There is
very limited literature regarding the incidence, presentation symptoms, diagnostic modalities, treatment and outcomes of LMM with these
novel therapies from LMICs. Over the last 5 years, we have reported four randomised studies in NSCLC from India [9–12]. These studies
have contributed towards the improvement in outcomes of NSCLC. In view of the limited data on LMM from NSCLC from LMIC, we carried
out this post-hoc analysis on the data obtained from these four prospective randomised studies, with a key objective to estimate the median
OS in patients who develop LMM after diagnosis of NSCLC. The other objectives were to estimate the incidence of LMM, risk factors, clinical
presentation and pattern of treatment.
Materials and methods
Paent selecon
This was a post-hoc analysis on the data collected from the four prospective randomised controlled trials conducted on NSCLC patients at a
single centre in the last 10 years. All the four studies have been published and the detailed study protocols are available online.
These trials included the following:
1. PatientswhohadEGFRsensitisingmutationsenrolledina1:1fashioneitherinthepemetrexedversusgefitinibtrial(Pem-Gefstudy,
February 2012 and April 2016) [9] or gefitinib versus gefitinib plus pemetrexed and carboplatin trial (Gef versus Gef+C study, August
2016 and August 2018) [10]. The primary end point was progression-free survival (PFS) with OS being a key secondary end point in
both studies.
2. Patientswhowerediagnosedwithsquamouscellcarcinomaofthelungswereenrolledina1:1fashioninthelow-dosegemcitabine
with platinum versus standard-dose gemcitabine with the platinum trial (May 2013–March 2018) [11]. OS was the primary end point.
3. Patientswithadenocarcinomawhowereenrolledinthepemetrexedversuserlotinibmaintenancetrial(November2014–March2017)
with quality of life being the primary end point [12].
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 3
Data collecon
The data were extracted from the trial database of the four aforementioned prospective trials. The demographic details at enrollment in
the trial and at LMM, driver mutation status, brain metastasis status and treatment details were extracted. In addition, clinical presentation,
diagnostic details, treatment, response assessment and the outcomes for LMM were extracted.
Outcome variable
The occurrence of LMM was defined as positive cerebrospinal fluid (CSF) cytology or CSF sampling performed only when symptomatic.
Contrast-enhanced magnetic resonance imaging (MRI) with gadolinium contrast features was consistent with the diagnosis of LMM [11]. CSF
cytology was carried out in 17 patients and MRI was carried out in 27 patients. CSF cytology was especially carried out in all symptomatic
patients who were eligible for intrathecal therapy. These neuroimaging features were the unequivocal presence of leptomeningeal nodules or
enhancement. The response to LMM was assessed in accordance with the RANO working group proposal [12]. OS was defined as the time
in months from the date of diagnosis of LMM to the date of death.
Stascal analysis
SPSS version 17 and RStudio version 3.1 were used for analysis. Descriptive statistics were carried out. The incidence of LMM was calculated
using a competing risk analysis. Death due to any other cause before developing LMM was considered as competing. The factors affecting
LMM development were identified using binary logistic regression analysis. The factors tested were age, performance status on the Eastern
Oncology Cooperative Group (ECOG PS) scale, driver mutation status and previous brain metastasis. OS was estimated using the Kaplan–
Meier analysis. COX regression analysis was used to identify factors affecting OS. p-value of 0.05 was considered significant.
Results
Incidence of LMM
From our database of 1148 patients, we identified 36 (0.031%; 95%CI: 0.022–0.043) patients of NSCLC who developed LMM in the
past decade. The incidence of LMM is shown in Figure 1. The median time to develop LMM was 14.92 months (interquartile range (IQR):
7.7–21.84). The risk factors for development of LMM are shown in Table 1.
Baseline characteriscs in paents with LMM
Data on baseline patient characteristics at development of LMM are given in Table 2. There were 22 males (61.1%) and 14 females (38.9%)
intheanalysis.Therewere23patients(63.9%)whowerebelowtheageof60(non-elderlygroup)and13(36.1%)were≥60years(elderly
group). Twenty-four of the 36 patients (66.7%) had developed LMM after one line of therapy, while 6 patients (16.7%) received 3 or more
lines of therapy.
Clinical features
Data on symptoms at presentation were available for 31 out of 36 patients (86.1%). Majority of the patients (15 of the 31 evaluable patients
(48.4%)) presented with symptoms of altered higher mental functions or seizures. Full data on symptoms at presentation is presented in
Table 3. The diagnosis of LMM was made by CSF analysis alone in 10 patients (27.8%), while imaging was performed in 18 patients (50%).
Diagnosis by both CSF analysis and imaging was made in 8 patients (22.2%). Only 1 of the 18 patients (5.5%) who were diagnosed by CSF
analysis required 2 lumbar punctures. Systemic progression was present in 20 out of 36 patients (55.6%) of NSCLC at the time of diagnosis
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 4
of LMM out of which 10 patients (20%) had brain progression. The median time for the development of LMM was 14.92 months (IQR:
7.7–21.84). Out of the 36 patients, 10 patients (27.8%) had brain progression or new-onset brain metastasis. The details of the diagnosis of
LMM are incorporated and presented in Table 3.
Figure 1. Incidence of leptomeningeal metastasis in the whole cohort. The green line depicts the incidence of leptomeningeal metastasis and the red line
depicts the occurrence of a compeng event – death.
Table 1. Factors predicting the development of leptomeningeal metastasis.
Variable Variable factors Leptomeningeal metastasis (%) OR (95%CI) p-value
Age Elderly (n = 439) 12 (2.7) 0.89 (0.44–1.8) 0.745
Non-elderly (n = 709) 24 (3.4) Reference
Gender Male (n = 744) 23 (3.1) 0.567 (0.283–1.135) 0.109
Female (n = 404) 13 (3.2) Reference
ECOG PS ECOG PS 0-1 (n = 1,033) 30 (2.9) 0.674 (0.269–1.687) 0.399
ECOG PS 2 (n = 115) 8 (5.2) Reference
Histopathology Non-squamous (n = 858) 35 (4.1%) 2.731 (0.32–23.318) 0.359
Squamous (n = 290) 1 (0.3%) Reference
Driver mutation status Present (n = 658) 31 (4.7) Reference 0.064
Absent (n = 490) 5 (1) 0.379 (0.136–1.058)
Brain metastasis Present (n = 153) 10 (6.5) Reference 0.035
Absent (n = 995) 26 (2.6) 0.45 (0.214–0.946)
ECOG PS, Eastern Oncology Cooperative Group performance status; OR, odds ratio; CI, confidence interval.
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 5
Table 2. Patient characteristics.
Variable Value (n = 36)
Age
Elderly
Non-Elderly
13 (36%)
23 (64%)
Gender
Male
Female
22 (61%)
14 (39%)
ECOG PS
0–1
2–3
29 (81%)
7 (19%)
Histology
Squamous
Non-squamous
1 (2.8%)
35 (97.2%)
Driver Mutation
EGFR mutation
Exon 19 deletion
Exon 21 L858R
Others
31 (86.1%)
19 (61.3%)
10 (32.3%)
2 ( 6.4%)
Brain metastasis
Yes
No
9 (25%)
27 (75%)
Treatment
Twenty-three patients out of the 36 (63.9%) patients with leptomeningeal metastasis received some form of treatment. Details are pre-
sented in Table 3. Out of the 36 patients, 6 received chemotherapy (16.7%) for leptomeningeal progression. Eighteen out of 31 EGFR
mutation-positive patients (58.1%) received first-generation TKIs; 7 received high-dose TKI (erlotinib/gefitinib) (38.9%) and the rest stan-
dard dose (61.1%). Only 6 (19.4%) out of 31 EGFR-mutated patients received osimertinib. Intrathecal treatment was administered to
11 of the 36 patients (30.6%) of which 4 received triple intrathecal therapy (36.4%) (methotrexate, cytarabine and hydrocortisone) and
7 received single-agent methotrexate (63.6%). Patients with objective responses were treated with osimertinib either with standard-dose
or high-dose intrathecal therapy. Patients with a stable disease were commonly treated with a combination of intrathecal therapy, TKI
(high-dose erlotinib or gefitinib) and chemotherapy (most commonly combining TKI and intrathecal therapy). The details of the responses
to intrathecal therapy are highlighted in Table 4.
Response
Response assessment was conducted in all 23 patients (63.9%) who were treated. Response was seen in 3 patients (13%), stable disease in
11 (47.8%) and in the remaining 9 (39.2%) had a progressive or refractory disease.
Overall survival
The median OS after development of LM was 61 days (95% CI: 38.95–83.05) (Figure 2). None of the tested factors, including age (Hazard
ratio = 0.829; 95% CI: 0.383–1.792, p-value = 0.633), ECOG PS (Hazard ratio = 0.745; 95% CI: 0.306–1.813, p-value = 0.517), driver
mutation status (Hazard ratio = 2.188, 95% CI: 0.756–6.336, p-value = 0.149), presence of brain metastasis (Hazard ratio = 1.432, 95%
CI: 0.609–3.365, p-value = 0.411) or presence of positive CSF cytology (Hazard ratio = 1.026; 95% CI: 0.455–2.315, p-value = 0.95), had
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 6
an impact on OS. The median OS in driver mutated patients was 66 days (95% CI: 14.74–117.26) versus 51 days (95% CI: 14.5–87.5)
(p-value = 0.201) in non-driver mutated patients. Among the driver mutated patients, patients treated with osimertinib had a median OS
of 245 days (95% CI: 215.48–274.52) versus 52 days (95% CI: 22.62–81.38) (p-value = 0.327) for those without.
Table 3. Clinical features, diagnosis and treatment details.
Variable Value (n = 36)
Symptoms
Altered higher mental functions(memory defects/altered
sensorium) and seizures
Cerebrovascular accident like episodes/visual disturbances
Headache and dizziness
15 (48.4%)
6 (19.4%)
10 (32.2%)
Diagnosis
CSF analysis
Imaging
CSF analysis and Imaging
10 (27.8%)
18 (50%)
8 (22.2%)
Number of other sites of metastasis
1
2
3
5 (13.8%)
21(58.3%)
10 (27.8%)
Brain metastasis prior to leptomeningeal metastasis
Yes
No 9 (25%)
27 (75%)
Number of previous lines of therapy
1
2
3
24 (66.6%)
6 (16.7%)
6 (16.7%)
Brain metastasis or progression at the time of
leptomeningeal metastasis
Yes
No
10 (27.8%)
26 (72.2%)
Intrathecal therapy
Methotrexate
Triple intrathecal therapy
11 (47.8%)
7 (63.6%)
4 (36.45)
Chemotherapy
Pemetrexed
Docetaxel
Paclitaxel
6 (26%)
2 (33.3%)
3 (50%)
1 (16.7%)
Tyrosine kinase inhibitor
Erlotinib
High
Standard
Gefitinib
High
Standard
Osimertinib
High
Standard
18 (78.2%)
3 (16.7%)
-
3 (33.3%)
6 (66.7%)
1 (16.7%)
5 (83.3%)
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 7
Table 4. Responses in patients receiving intrathecal therapy.
Response Number of patients Intrathecal therapy
(Number of patients)
Complete response 1 Intrathecal methotrexate (1)
Partial response 2 Intrathecal therapy (2)
Triple intrathecal therapy (1)
Intrathecal methotrexate (1)
Stable disease 11 Intrathecal therapy (6)
Triple intrathecal therapy (2)
Intrathecal methotrexate (4)
No intrathecal therapy (5)
Progressive disease 17 Intrathecal therapy (2)
Triple intrathecal therapy (0)
Intrathecal methotrexate (2)
No intrathecal therapy (15)
Figure 2. OS graph. OS in days with its 95% condence interval is depicted. The median OS value is depicted with dashed lines.
Discussion
LMM is a devastating complication associated with solid malignancies with limited treatment options and poor OS. To the best of authors’
knowledge, this is one of the few studies that has retrospectively looked into four prospective single-centre trials on LMM in NSCLC. The
incidence of LMM in the current study is 0.03%. This is much lower than the incidence reported in multiple studies which ranges between
3% and 9% [4, 5]. The question is why was the incidence of leptomeningeal metastasis low in our study. The reason may be manifold. Firstly,
evaluation of the craniospinal axis was not carried out routinely. It was performed by physicians only when brain metastasis or LMM were
suspected. Hence, asymptomatic LMM was missed. This would have led to falsely low rates of LMM. Secondly, we used strict criteria for
diagnosis of LMM which were either cytological positivity or radiological evidence. The diagnosis of LMM based on biochemical or clinical
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 8
parameters was avoided even though these recommendations are provided by the NCCN and the ESMO-EANO group [13, 14]. Thirdly,
access to second and third-generation TKIs and immunotherapy was restricted in our studies. Therefore, the 18-month OS rate in EGFR
mutation-positive patients in India is between 48.7 and 74.3% [9, 10] as opposed to the 18-month OS rate with third-generation TKIs of 85%
[15, 16]. Probably because of this, patients died due to extracranial progression (competing causes), thus limiting the probability of progres-
sion to LMM. Fourthly, our data were from a prospective cohort of four studies. As opposed to this, the incidence data in most of the studies
has come from retrospective studies with its likelihood of selection bias. Lastly, there might be a lower occurrence of LMM in Indian patients.
Time to develop LMM in the current study was 14.92 months. This is similar to the time to development seen in other studies. In a ret-
rospective analysis from Kuiper et al [17], time to development of leptomeningeal metastasis was 13·3 months. In our study, the trend of
occurrence of LMM was more in driver mutation-driven patients, which is in concurrence with previous studies. In a study by Li et al [6],
the incidence of LMM was found to be 9.4% in patients with EGFR mutations versus 0.7% in wild-type EGFR patients (p < 0·001). The
higher occurrence of LMM in patients with brain metastasis suggests that it is important to consider the occurrence of LMM in patients
with brain metastasis on follow-up [18].
The clinical features seen in our study had a predominance (nearly half) of higher mental function disturbance and seizures. This is in line
with the data published by Pan et al where adenocarcinoma-related LMM mainly leads to higher mental function symptoms [19]. Another
interesting aspect of the data was presentation of LMM with cerebrovascular accident-like features. This suggests that LMM probably should
be a provisional diagnosis when CVA-like symptoms are seen in NSCLC patients.
Nearly 40% of the patients in the current study did not receive cancer-directed treatment at LMM. This is in line with the data across the
world [20]. The outcomes of patients with poor performance status, fixed extensive neurological deficits or with extracranial progression are
dismal [21]. Hence, patients with these features are not treated routinely at our centre. The variable use of intrathecal therapies with sys-
temic therapies seen in the study is in concurrence with the limited efficacy of intrathecal therapies seen in the literature [1, 22]. There is no
evidence of improvement in OS or PFS with the use of intrathecal therapies [1, 22]. The median OS after development of LMM was around
2 months, which is consistent with that quoted in the literature [21–23]. As per our study, the use of intrathecal therapy had some form of
response among patients as compared to those who did not receive intrathecal therapy.
Our results are inferior to those reported in studies focussing on novel agents in driver mutated patients. The recently published BLOOM
study has shown promising results with the use of high-dose osimertinib (80 mg BID) in EGFR-mutated NSCLC with LMM [25]. In our study
also there was a trend towards higher OS in patients receiving osimertinib. However, the numbers were few (6 out of 31) to have adequate
power for a statistical analysis. Thus, our data along with the literature suggests that non-driver mutated patients have poor prognosis and
even driver mutated patients would have similar prognosis if exposure to third-generation novel agents is not possible. The outcomes of
EGFR-mutated NSCLC treated with first-generation tyrosine kinase inhibitors can be improved by administering either VEGF inhibitors [26]
or with the addition of chemotherapy [10] or alternatively by substituting it with novel third-generation agents [18]. Moreover, the results
imply that in patients with LMM, the use of agents like osimertinib is warranted.
The study limitations are that it was carried out among the Indian population and in a single centre. Access to third-generation therapies and
immunotherapy was limited. However, this is also a strength as it is a pointer towards the likely outcomes in LMICs where access to these
agents are limited. The strength of the study is the strict criteria used for LMM diagnosis and the radiology review for confirmation of the
presence of LMM.
Conclusion
The incidence of LMM is low in the Indian population. In our study, there was no single factor which impacted survival in patients who developed
LMM. This suggests that the overall prognosis is poor in patients with LMM where access to newer therapeutic modalities is limited.
Funding
No funding was received for this study.
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 9
Conicts of interest
The authors declare that they have no conflicts of interest.
Authors’ contribuons
Vijay Patil and Kumar Prabhash – concept, design, execution, data collection, analysis, interpretation, first draft and final draft.
Vanita Noronha – execution, data collection, interpretation, first draft and final draft.
Dilip Harindran Vallathol – concept, data collection, interpretation, first draft and final draft.
Nandini Menon, Abhishek Mahajan, Amit Janu and Nilendu Purandare – execution, data collection and final draft
References
1. Vallathol DH, Patil VM, and Noronha V, et al (2020) Leptomeningeal metastasis from extracranial solid tumors Cancer Res Stat Treat 3(2)
254 https://doi.org/10.4103/CRST.CRST_38_20
2. Soffietti R, Abacioglu U, and Baumert B, et al (2017) Diagnosis and treatment of brain metastases from solid tumors: guidelines from
the European Association of Neuro-Oncology (EANO) Neuro Oncol 19(2) 162–174 https://doi.org/10.1093/neuonc/now241 PMID:
28391295 PMCID: 5620494
3. Le Rhun E, Preusser M, and van den Bent M, et al (2019) How we treat patients with leptomeningeal metastases ESMO Open 4(Suppl 2)
e000507 https://doi.org/10.1136/esmoopen-2019-000507 PMID: 31231573 PMCID: 6555600
4. Cheng H and Perez-Soler R (2018) Leptomeningeal metastases in non-small-cell lung cancer Lancet Oncol 19(1) e43–e55 https://doi.
org/10.1016/S1470-2045(17)30689-7 PMID: 29304362
5. Remon J, Le Rhun E, and Besse B (2017) Leptomeningeal carcinomatosis in non-small cell lung cancer patients: a continuing challenge
in the personalized treatment era Cancer Treat Rev 53 128–137 https://doi.org/10.1016/j.ctrv.2016.12.006 PMID: 28110254
6. Li Y-S, Jiang B-Y, and Yang J-J, et al (2016) Leptomeningeal metastases in patients with NSCLC with EGFR mutations J Thorac Oncol
11(11) 1962–1969 https://doi.org/10.1016/j.jtho.2016.06.029 PMID: 27539328
7. Liao BC, Lee JH, and Lin CC, et al (2015) Epidermal growth factor receptor tyrosine kinase inhibitors for non-small-cell lung cancer
patients with leptomeningeal carcinomatosis J Thorac Oncol 10(12) 1754–1761 https://doi.org/10.1097/JTO.0000000000000669
PMID: 26334749
8. Zhao J, Chen M, and Zhong W, et al (2013) Cerebrospinal fluid concentrations of gefitinib in patients with lung adenocarcinoma Clin
Lung Cancer 14(2) 188–193 https://doi.org/10.1016/j.cllc.2012.06.004
9. Patil VM, Noronha V, and Joshi A, et al (2017) Phase III study of gefitinib or pemetrexed with carboplatin in EGFR-mutated advanced
lung adenocarcinoma ESMO Open 2(1) e000168 https://doi.org/10.1136/esmoopen-2017-000168 PMID: 28761735 PMCID: 5519810
10. Noronha V, Patil VM, and Joshi A, et al (2019) Gefitinib versus gefitinib plus pemetrexed and carboplatin chemotherapy in EGFR-
mutated lung cancer J Clin Oncol Published online August 14 JCO1901154
11. Patil V, Noronha V, and Joshi A, et al (2019) Phase III non-inferiority study evaluating efficacy and safety of low dose gemcitabine
compared to standard dose gemcitabine with platinum in advanced squamous lung cancer EClinicalMedicine 9 19–25 https://doi.
org/10.1016/j.eclinm.2019.03.011 PMID: 31143878 PMCID: 6510888
Research
ecancer 2022, 16:1414; www.ecancer.org; DOI: https://doi.org/10.3332/ecancer.2022.1414 10
12. Patil V, Joshi A, and Noronha V, et al (2019) Randomized phase 3 open label study of quality of life of patients on Pemetrexed versus
Erlotinib as maintenance therapy for advanced non squamous non EGFR mutated non small cell lung cancer Oncotarget 10(59) 6297–
6307 https://doi.org/10.18632/oncotarget.27214 PMID: 31695838 PMCID: 6824869
13. Le Rhun E, Weller M, and Brandsma D, et al (2017) EANO-ESMO clinical practice guidelines for diagnosis, treatment and follow-up
of patients with leptomeningeal metastasis from solid tumours Ann Oncol 28(suppl_4) iv84–iv99 https://doi.org/10.1093/annonc/
mdx221 PMID: 28881917
14. Chamberlain M, Junck L, and Brandsma D, et al (2017) Leptomeningeal metastases: a RANO proposal for response criteria Neuro Oncol
19(4) 484–492 PMID: 28039364 PMCID: 5464328
15. Soria J-C, Ohe Y, and Vansteenkiste J, et al (2018) Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer N Engl
J Med 378(2) 113–125 https://doi.org/10.1056/NEJMoa1713137
16. Ramalingam SS, Vansteenkiste J, and Planchard D, et al (2020) Overall survival with osimertinib in untreated, EGFR-mutated advanced
NSCLC N Engl J Med 382(1) 41–50 https://doi.org/10.1056/NEJMoa1913662
17. Kuiper JL, Hendriks LE, and van der Wekken AJ, et al (2015) Treatment and survival of patients with EGFR-mutated non-small cell
lung cancer and leptomeningeal metastasis: a retrospective cohort analysis Lung Cancer 89(3) 255–261 https://doi.org/10.1016/j.
lungcan.2015.05.023 PMID: 26117231
18. Chowdhary S and Chamberlain M (2005) Leptomeningeal metastases: current concepts and management guidelines J Natl Compr Canc
Netw 3(5) 693–703 https://doi.org/10.6004/jnccn.2005.0039 PMID: 16194457
19. Pan Z, Yang G, and He H, et al (2018) Leptomeningeal metastasis from solid tumors: clinical features and its diagnostic implication Sci
Rep 8(1) 10445 https://doi.org/10.1038/s41598-018-28662-w PMID: 29992998 PMCID: 6041294
20. Morris PG, Reiner AS, and Szenberg OR, et al (2012) Leptomeningeal metastasis from non-small cell lung cancer: survival and the
impact of whole brain radiotherapy J Thorac Oncol 7(2) 382–385 https://doi.org/10.1097/JTO.0b013e3182398e4f
21. Grossman SA, Finkelstein DM, and Ruckdeschel JC, et al (1993) Randomized prospective comparison of intraventricular methotrex-
ate and thiotepa in patients with previously untreated neoplastic meningitis. Eastern Cooperative Oncology Group J Clin Oncol 11(3)
561–569 https://doi.org/10.1200/JCO.1993.11.3.561 PMID: 8445432
22. Boogerd W, van den Bent MJ, and Koehler PJ, et al (2004) The relevance of intraventricular chemotherapy for leptomeningeal metasta-
sis in breast cancer: a randomised study Eur J Cancer 40(18) 2726–2733 https://doi.org/10.1016/j.ejca.2004.08.012 PMID: 15571954
23. Hitchins RN, Bell DR, and Woods RL, et al (1987) A prospective randomized trial of single-agent versus combination chemotherapy in
meningeal carcinomatosis J Clin Oncol 5(10) 1655–1662 https://doi.org/10.1200/JCO.1987.5.10.1655 PMID: 3309199
24. Glantz MJ, Jaeckle KA, and Chamberlain MC, et al (1999) A randomized controlled trial comparing intrathecal sustained-release cytara-
bine (DepoCyt) to intrathecal methotrexate in patients with neoplastic meningitis from solid tumors Clin Cancer Res 5(11) 3394–3402
PMID: 10589750
25. Yang JCH, Kim S-W, and Kim D-W, et al (2020) Osimertinib in patients with epidermal growth factor receptor mutation-positive
non-small-cell lung cancer and leptomeningeal metastases: the BLOOM study J Clin Oncol 38(6) 538–547 https://doi.org/10.1200/
JCO.19.00457 PMCID: 7030895
26. Chen Z, Wei J, and Ma X, et al (2020) Efficacy of EGFR-TKIs with or without angiogenesis inhibitors in advanced non-small-cell lung
cancer: a systematic review and meta-analysis J Cancer 11(3) 686–695 https://doi.org/10.7150/jca.34957 PMID: 31942192 PMCID:
6959046
... In a LMC experience reported from Kochi (State of Kerala), the commonest site leading to LMC was breast (45%) followed by lung cancer (35%). 21 Other large experiences from India (>1100 patients) by Patil and colleagues 22 and Abraham and colleagues, 23 were in lung cancer and breast cancer respectively. A significant proportion of patients (>50%) have presented in ECOG PS 2-4 state. ...
... In a large experience from prospective studies in NSCLC, the symptoms of leptomeningeal disease were symptoms of altered higher mental functions (48.4%) or seizures and Headache and dizziness (32.2%). 22 Similar experience was published from Kochi, where the commonest presenting features were headache, vomiting, loss of consciousness, cranial nerve palsies and seizures. 21 The treatment pattern suggested that a significant proportion of patients were offered the best supportive care in our study. ...
... In a LMC experience reported from Kochi (State of Kerala), the commonest site leading to LMC was breast (45%) followed by lung cancer (35%). 21 Other large experiences from India (>1100 patients) by Patil and colleagues 22 and Abraham and colleagues, 23 were in lung cancer and breast cancer respectively. A significant proportion of patients (>50%) have presented in ECOG PS 2-4 state. ...
... In a large experience from prospective studies in NSCLC, the symptoms of leptomeningeal disease were symptoms of altered higher mental functions (48.4%) or seizures and Headache and dizziness (32.2%). 22 Similar experience was published from Kochi, where the commonest presenting features were headache, vomiting, loss of consciousness, cranial nerve palsies and seizures. 21 The treatment pattern suggested that a significant proportion of patients were offered the best supportive care in our study. ...
Article
Increased survival (due to the use of targeted therapies based on genomic profiling) has resulted in the increased incidence of brain metastasis during the course of disease, and thus, made it essential to have proper imaging guidelines in place for brain metastasis from non-small-cell lung cancer (NSCLC). Brain parenchymal metastases can have varied imaging appearances, and it is pertinent to be aware of the various molecular risk factors for brain metastasis from NSCLC along with their suggestive imaging appearances, so as to identify them early. Leptomeningeal metastasis requires additional imaging of the spine and an early cerebrospinal fluid (CSF) analysis. Differentiation of post-therapy change from recurrence on imaging has a bearing on the management, hence the need for its awareness. This article will provide in-depth literature review of the epidemiology, aetiopathogenesis, screening, detection, diagnosis, post-therapy imaging, and implications regarding the management of brain metastasis from NSCLC. In addition, we will also briefly highlight the role of artificial intelligence (AI) in brain metastasis screening.
Article
Full-text available
In the present study, we evaluated the efficacy and safety of epidermal growth factor receptor tyrosine kinases (EGFR-TKIs) combined with or without angiogenesis inhibitors in advanced non-small-cell lung cancer (NSCLC). We searched published randomized controlled trials (RCTs) comparing EGFR-TKIs with and without angiogenesis inhibitors for the treatment of advanced NSCLC. PubMed, EMBASE, PMC, the American Society of Clinical Oncology (ASCO) and the European Society of Medical Oncology (ESMO) databases were searched. The extracted data on progression-free survival (PFS) and overall survival (OS) were measured in terms of hazard ratios (HRs) and corresponding 95% confidence intervals (CIs). In addition, odds ratios (ORs) and corresponding 95% CIs were pooled for objective response rate (ORR) and disease control rate (DCR). Risk ratios (RRs) and corresponding 95% CIs were pooled for risk of adverse events (AEs). EGFR-TKIs combined with angiogenesis inhibitors showed significant improvements in PFS (HR 0.72, 95% CI 0.61-0.84, P <0.0001), ORR (OR 1.51, 95% CI 1.17-1.97, P=0.002) and DCR (OR 1.49, 95% CI 1.24-1.81, P<0.0001) compared with EGFR-TKIs combined with placebo. However, EGFR-TKIs combined with angiogenesis inhibitors failed to improve OS (HR 0.94, 95% CI 0.84-1.05, P = 0.26). In addition, diarrhea, hypertension, thrombocytopenia, neutropenia, fatigue, rash, and dermatitis acneiform were significantly increased in patients treated with angiogenesis inhibitors. Thus, EGFR-TKIs combined with angiogenesis inhibitors were superior to EGFR-TKIs alone in advanced NSCLC due to their effects on PFS, ORR and DCR, but the increased incidence of AEs had an influence on the tolerability of this combination therapy.
Article
Full-text available
PURPOSE In this phase I study (BLOOM), osimertinib, a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), was evaluated in patients with leptomeningeal metastases (LMs) from EGFR-mutated (EGFRm) advanced non–small-cell lung cancer (NSCLC) whose disease had progressed on previous EGFR-TKI therapy. PATIENTS AND METHODS Patients with cytologically confirmed LM received osimertinib 160 mg once daily. Objectives were to assess confirmed objective response rate (ORR), duration of response (DoR), progression-free survival (PFS), overall survival (OS), pharmacokinetics (PK), and safety. Additional efficacy evaluations included changes from baseline in CSF cytology and neurologic examination. Measurable lesions were assessed by investigator according to RECIST version 1.1. LMs were assessed by neuroradiologic blinded central independent review (BICR) according to Response Assessment in Neuro-Oncology LM radiologic criteria and by investigator. RESULTS Forty-one patients were enrolled. LM ORR and DoR by neuroradiologic BICR were 62% (95% CI, 45% to 78%) and 15.2 months (95% CI, 7.5 to 17.5 months), respectively. Overall, ORR by investigator was 41% (95% CI, 26% to 58%), and median DoR was 8.3 months (95% CI, 5.6 to 16.5 months). Median investigator-assessed PFS was 8.6 months (95% CI, 5.4 to 13.7 months) with 78% maturity; median OS was 11.0 months (95% CI, 8.0 to 18.0 months) with 68% maturity. CSF tumor cell clearance was confirmed in 11 (28%; 95% CI, 15% to 44%) of 40 patients. Neurologic function was improved in 12 (57%) of 21 patients with an abnormal assessment at baseline. The adverse event and PK profiles were consistent with previous reports for osimertinib. CONCLUSION Osimertinib showed meaningful therapeutic efficacy in the CNS and a manageable safety profile at 160 mg once daily in patients with EGFRm NSCLC and LM.
Article
Full-text available
Background: We planned to compare pemetrexed maintenance with erlotinib maintenance in non squamous non Epidermal Growth Factor Receptor (EGFR) mutated non small cell lung cancer (NSCLC). The null hypothesis for this study was that there would be no difference in quality of life (QOL) between pemetrexed and erlotinib maintenance. Results: The QL2 scores at 3 months were 63.35 (SD 24.99) in pemetrexed arm and 63.01(SD 23.04) in erlotinib arm (p-0.793). Except in 1 domain, the scores were statistically similar between the 2 arms. In the domain of diarrhea, the score was higher as expected in the erlotinib arm (p-0.048). The median progression free survival was 4.5 months (95%CI 4.1–4.9 months) in pemetrexed arm versus 4.5 months (95%CI 3.8–5.2 months) in erlotinib arm (p-0.94). The median overall survival was 16.6 months (15.2–17.9 months) in pemetrexed arm versus 18.3 months (95% CI 13.75–22.91 months) in erlotinib arm (p-0.49). Methods: The study was an open label, single centre, parallel, phase 3 randomized study with 1:1 randomization between maintenance pemetrexed arm and erlotinib arm. Adult patients (age > or = 18 years), with non squamous EGFR mutation, treated with first line palliative therapy, with non progressive disease post 4–6 cycles of pemetrexed-carboplatin were randomized. Primary outcome was change in the score of QOL (Global health status {QL2}) at 3 months. We estimated that with 200 patients, the study had 80% power to detect a significant difference between the two groups in the change in the global health status score at 3 months with an alpha error of 5%, with an effect size of 0.3 SD. Conclusions: Maintenance pemetrexed post pemetrexed-platinum chemotherapy fails to improve QOL or time to event outcomes over maintenance erlotinib in EGFR mutation negative NSCLC.
Article
Full-text available
The goal of treatment of leptomeningeal metastasis is to improve survival and to maintain quality of life by delaying neurological deterioration. Tumour-specific therapeutic options include intrathecal pharmacotherapy, systemic pharmacotherapy and focal radiotherapy. Recently, improvement of leptomeningeal disease–related progression-free survival by adding intrathecal liposomal cytarabine to systemic treatment versus systemic treatment alone has been observed in a randomised phase III trial for patients with breast cancer with newly diagnosed leptomeningeal metastasis. Safety and efficacy of intrathecal administration of new agents such as trastuzumab are under evaluation. Systemic therapy using targeted agents and immunotherapy has also improved outcome in patients with brain metastasis, and its emerging role in the management of leptomeningeal metastasis needs to better studied in prospective series. Focal radiotherapy is commonly indicated for the treatment of macroscopic disease such as meningeal nodules or clinically symptomatic central nervous system structures, for example, base of skull with cranial nerve involvement or cauda equine syndrome. The role of whole brain radiotherapy is decreasing. An individualised combination of different therapeutic options should be used considering the presentation of leptomeningeal metastasis, as well as the histological and molecular tumour characteristics, the presence of concomitant brain and systemic metastases, and prior cancer-directed treatments.
Article
Full-text available
In this study, we examined the characteristics and aimed to increase the knowledge of clinical features of leptomeningeal metastasis (LM). The clinical data, including initial diagnosis and treatment of primary tumor, clinical manifestations, neuroimaging findings, cerebrospinal fluid (CSF) examination, were analyzed. For the patients with adenocarcinoma/breast cancer, the incidence of cranial lesions and cranial nerve paralysis was obviously higher than patients with small cell lung cancer. Whereas, the incidence of involvement of intravertebral canal was obviously lower than that of small cell lung cancer. Patients with adenocarcinoma/breast cancer showed more incidence of leptomeningeal enhancement compared to those with small cell lung cancer. Persistent severe headache was noticed in those with squamous carcinoma, and usually showed absence of abnormally LM-related neuroimaging and CSF cytological findings, which resulted in a challenge in the diagnosis of LM from squamous carcinoma. Patients with different primary tumors showed differential clinical features. Significant differences were observed in clinical features between patients with adenocarcinoma/breast cancer and small cell lung cancer. Our study contributes to the understanding of clinical characteristics of LM, and contributes to improvement of LM diagnosis in clinical practice.
Article
Background: Osimertinib is a third-generation, irreversible tyrosine kinase inhibitor of the epidermal growth factor receptor (EGFR-TKI) that selectively inhibits both EGFR-TKI-sensitizing and EGFR T790M resistance mutations. A phase 3 trial compared first-line osimertinib with other EGFR-TKIs in patients with EGFR mutation-positive advanced non-small-cell lung cancer (NSCLC). The trial showed longer progression-free survival with osimertinib than with the comparator EGFR-TKIs (hazard ratio for disease progression or death, 0.46). Data from the final analysis of overall survival have not been reported. Methods: In this trial, we randomly assigned 556 patients with previously untreated advanced NSCLC with an EGFR mutation (exon 19 deletion or L858R allele) in a 1:1 ratio to receive either osimertinib (80 mg once daily) or one of two other EGFR-TKIs (gefitinib at a dose of 250 mg once daily or erlotinib at a dose of 150 mg once daily, with patients receiving these drugs combined in a single comparator group). Overall survival was a secondary end point. Results: The median overall survival was 38.6 months (95% confidence interval [CI], 34.5 to 41.8) in the osimertinib group and 31.8 months (95% CI, 26.6 to 36.0) in the comparator group (hazard ratio for death, 0.80; 95.05% CI, 0.64 to 1.00; P = 0.046). At 3 years, 79 of 279 patients (28%) in the osimertinib group and 26 of 277 (9%) in the comparator group were continuing to receive a trial regimen; the median exposure was 20.7 months and 11.5 months, respectively. Adverse events of grade 3 or higher were reported in 42% of the patients in the osimertinib group and in 47% of those in the comparator group. Conclusions: Among patients with previously untreated advanced NSCLC with an EGFR mutation, those who received osimertinib had longer overall survival than those who received a comparator EGFR-TKI. The safety profile for osimertinib was similar to that of the comparator EGFR-TKIs, despite a longer duration of exposure in the osimertinib group. (Funded by AstraZeneca; FLAURA ClinicalTrials.gov number, NCT02296125.).
Article
Purpose: Standard first-line therapy for EGFR-mutant advanced non-small-cell lung cancer (NSCLC) is an epidermal growth factor receptor (EGFR)-directed oral tyrosine kinase inhibitor. Adding pemetrexed and carboplatin chemotherapy to an oral tyrosine kinase inhibitor may improve outcomes. Patients and methods: This was a phase III randomized trial in patients with advanced NSCLC harboring an EGFR-sensitizing mutation and a performance status of 0 to 2 who were planned to receive first-line palliative therapy. Random assignment was 1:1 to gefitinib 250 mg orally per day (Gef) or gefitinib 250 mg orally per day plus pemetrexed 500 mg/m2 and carboplatin area under curve 5 intravenously every 3 weeks for four cycles, followed by maintenance pemetrexed (gefitinib plus chemotherapy [Gef+C]). The primary end point was progression-free survival (PFS); secondary end points included overall survival (OS), response rate, and toxicity. Results: Between 2016 and 2018, 350 patients were randomly assigned to Gef (n = 176) and Gef+C (n = 174). Twenty-one percent of patients had a performance status of 2, and 18% of patients had brain metastases. Median follow-up time was 17 months (range, 7 to 30 months). Radiologic response rates were 75% and 63% in the Gef+C and Gef arms, respectively (P = .01). Estimated median PFS was significantly longer with Gef+C than Gef (16 months [95% CI, 13.5 to 18.5 months] v 8 months [95% CI, 7.0 to 9.0 months], respectively; hazard ratio for disease progression or death, 0.51 [95% CI, 0.39 to 0.66]; P < .001). Estimated median OS was significantly longer with Gef+C than Gef (not reached v 17 months [95% CI, 13.5 to 20.5 months]; hazard ratio for death, 0.45 [95% CI, 0.31 to 0.65]; P < .001). Clinically relevant grade 3 or greater toxicities occurred in 51% and 25% of patients in the Gef+C and Gef arms, respectively (P < .001). Conclusion: Adding pemetrexed and carboplatin chemotherapy to gefitinib significantly prolonged PFS and OS but increased toxicity in patients with NSCLC.
Article
Background Prolonged infusion of low dose gemcitabine (PLDG) in combination with platinum has shown promising activity in terms of improved response rate and progression free survival (PFS); especially in squamous non-small cell lung cancer (NSCLC). Hence, we conducted a phase 3 randomized non-inferiority study with the primary objective of comparing the overall survival (OS) between PLDG and standard dose of gemcitabine with platinum. Methodology Adult subjects (age ≥ 18 years), with stages IIIB–IV, NSCLC (squamous) and ECOG performance status of ≤ 2 were randomized 1:1 into either carboplatin with standard dose gemcitabine (1000 mg/m² intravenous over 30 min, days 1 and 8) (STD-G arm) or carboplatin along with low dose gemcitabine (250 mg/m² intravenous over 6 h, days 1 and 8) (LOW-G arm) for a maximum of 6 cycles. Tumor response was assessed by RECIST criteria version 1.1 every 2 cycles till 6th cycle and thereafter at 2 monthly intervals till progression. The primary endpoint was overall survival. 308 patients were randomized, 155 in STD-G arm and 153 in LOW-G arm, respectively. Results The median overall survival in STD-G arm was 6.8 months (95%CI 5.3–8.5) versus 8.4 months (95%CI 7–10.3) in the LOW-G arm (HR-0.890 (90%CI 0.725–1.092). The results with per protocol analysis were in line with these results. There was no statistical difference in progression free survival (HR-0.949; 90%CI 0.867–1.280) and adverse event rate between the 2 arms. Conclusion This study suggests that PLDG is an alternative to the standard gemcitabine schedule in squamous NSCLC, and either of these can be selected subject to patient convenience.
Article
Leptomeningeal metastasis is a complication of advanced non-small-cell lung cancer (NSCLC). Diagnosis and monitoring of leptomeningeal metastasis are challenging, and are based on neurological, radiographic, and cerebrospinal fluid findings. Substantial progress has been made in several key aspects of management of leptomeningeal metastasis, including improved characterisation of the genetic profiles, generation of clinically relevant animal models, advances in cerebrospinal fluid liquid biopsy with improved cytology and genotyping analysis, and the development of therapeutic agents with greater CNS penetration. This Review discusses cumulative data on multiple treatment modalities with a particular focus on recent advances in molecularly targeted therapies in subtypes of patients with leptomeningeal metastasis from NSCLC. Future research is needed to further understand the biology of leptomeningeal metastasis and the mechanisms of resistance to treatment.