• 患者服务: 与癌共舞小助手
  • 微信号: yagw_help22

QQ登录

只需一步,快速开始

开启左侧

肺腺癌

    [复制链接]
211156 163 荷花池荒岛 发表于 2014-3-24 12:31:48 |
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:53:06 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-11 11:35 编辑

The Landscape of EGFR Pathways and Personalized Management of Non-small-cell Lung Cancer
http://www.medscape.com/viewarticle/740715_1

Abstract and Introduction

Abstract

Two classes of anti-EGF receptor (EGFR) agents, monoclonal anti-EGFR antibodies and small-molecule EGFR tyrosine kinase inhibitors, have been used for the treatment of non-small-cell lung cancer (NSCLC). However, only a subset of patients will benefit from EGFR-targeted therapy. The discovery of biomarkers that select the appropriate patients for the therapy and predict the responses to the therapy is urgently needed. Molecular genetic analyses provide new insights into EGFR pathway alterations and demonstrate promise for predicting the clinical outcome of patients with NSCLC. In this article, we summarize the latest available knowledge on the clinical impact of EGFR mutations, gene copy number, EGFR overexpression, phosphorylation expression and the alteration of the EGFR pathway downstream factors in predicting the response to EGFR-targeted therapy in NSCLC patients. The role of KRAS and BRAF mutations and ALK rearrangement in lung cancer-targeted therapy, are also reviewed.

Introduction

Non-small-cell lung cancer (NSCLC) accounts for approximately 80% of lung cancers and is one of the leading causes of cancer death in North America. It is often diagnosed at an advanced stage, with only 30–40% of metastatic NSCLC patients surviving for 12 months.[1–3]

Surgery is the most effective treatment for NSCLC; however, it is usually reserved for patients whose tumors are confined to the primary site and who have no or minimal lymph node involvement – a small portion of NSCLC cases. In addition, many patients who undergo curative surgery will later develop recurrence. Platinum-based chemotherapy is the mainstay of treatment in advanced or recurrent NSCLC, but the results of the treatment are far from encouraging.[1]

Approximately 85% of all primary lung cancers are NSCLCs, which are classified into three major histologic types: adenocarcinoma, squamous cell carcinoma and large-cell carcinoma.[4] Adenocarcinoma is the most frequent histologic type of NSCLC in both genders in many parts of the world (Figure 1). It accounts for approximately 40% of lung cancers and is usually found in the periphery of the lungs.[1] The most common histologic subtypes of adenocarcinoma are papillary (37%), acinar (30%), solid (25%) and bronchioloalveolar (7%).[5] Squamous cell carcinoma accounts for approximately 25–30% of all lung cancers and is often linked to a history of smoking. This cancer tends to occur in the hilar region of the lungs near the bronchus. Large-cell carcinoma accounts for approximately 10–15% of lung cancers and may appear in any part of the lung. It tends to grow and spread quickly and carries a poor prognosis.

Cigarette smoking remains the principal cause of lung cancer. It is estimated that 85–90% of all lung cancer patients have smoked cigarettes at some time.[6] As such, 87% of lung cancer deaths are thought to result from smoking. The US Environmental Protection Agency reports that radon is the second leading cause of lung cancer after cigarette smoking and is the leading environmental cause for nonsmokers. The risk of developing NSCLC from radon is much higher in people who smoke than in those who do not. Workplace exposure to asbestos fibers is another important, but less common, risk factor for lung cancer.[7–9]

In recent years, attention has turned to the role that the EGF receptor gene (EGFR) plays in tumorigenesis and its utility as a target for therapy. Biomarkers that can reliably predict which patients may benefit from anti-EGFR therapy are urgently needed. Pathologists will play a central role in the process to determine suitable testing and interpretation of the test results. In this article, we summarize the impact of EGFR alterations in predicting response to anti-EGFR therapies and discuss currently proposed technologies and their potential clinical implications.

Overview of EGFR & NSCLC

The EGFR and members of its family play an important role in carcinogenesis through their involvement in the modulation of cell proliferation, apoptosis, cell motility and neovascularization.[10] EGFR alterations have been implicated in the pathogenesis and progression of many malignancies.[11–14] Although the exact molecular pathways by which the mutant receptors lead to carcinogenesis are not completely understood, it is clear that mutant variants of EGFR have enhanced tyrosine kinase (TK) activity.

The presence of activating mutations in EGFR was initially reported in 2004.[12,15,16] Various groups also found amplification and overexpression of EGFR.[17–21] Clinical and pathological factors such as female gender, never having smoked, East Asian ethnicity and adenocarcinoma or bronchioloalveolar histology, correlated with objective responses to single-agent TK inhibitor (TKI) therapy in NSCLC and also with the presence of somatic EGFR mutations.[10,11,15,22,23] EGFR mutations rarely occurred in squamous cell carcinoma, large-cell carcinomas or adenocarcinomas with KRAS mutations.[5,12,15,20,22–31] Of great benefit to researchers in evaluating possible contributing mutations in NSCLC and acquired resistance to targeted therapies is the Catalogue of Somatic Mutations in Cancer (COSMIC[201]). COSMIC is designed to store and display somatic mutation information and related details relating to human cancers; it is a very useful tool for both researchers and clinicians. As more advanced molecular techniques reveal further molecular mutations, centralized databases such as COSMIC will allow clinicians and researchers to stay abreast of currently available information.

Lung adenocarcinomas frequently possess EGFR mutations and frequently exhibit increased EGFR copy number.[32] A study of 334 cases of lung adenocarcinomas using PCR-based assays to detect deletions within exon 19 and the L858R mutation in exon 21 of the EGFR gene found that 23% of these tumors contained a mutation. Of those, 71% were exon 19 deletions and 29% comprised the L858R mutation in exon 21.[32] In addition, EGFR amplification, defined as greater than five EGFR signals per nucleus by FISH, was detected in 52% of EGFR-mutated tumors, but in only 6% of those lacking the mutations. EGFR mutations were present in 26% of 86 bronchioloalveolar carcinomas.[24] It appears that EGFR mutations occur much less frequently in squamous cell carcinoma than in adenocarcinoma, with a reported incidence of 0–14%.[24,33] The third type of NSCLC, large-cell carcinoma, harbors EGFR mutations very rarely, if ever.[23,34] Marchetti et al. investigated a series of 31 large-cell carcinomas and found no EGFR mutations in any case.[24]

Motoi et al. reported that EGFR mutations are particularly frequent in adenocarcinoma of the papillary subtype.[5] In a group of NSCLC patients, 13 out of 36 (35%) papillary cancers harbored EGFR mutations, in contrast to three out of 63 nonpapillary cancers (5%). Kim et al. found that papillary subtype is a significant predictor of response to gefitinib in lung adenocarcinoma, although they did not link the incidence of response to the EGFR mutation status.[35]

The clinical implications of EGFR overexpression have been studied extensively, but the results are inconclusive thus far. Recent use of phosphor-specific antibody has facilitated analysis of the correlation between phosphorylation and EGFR mutation status. In a study of 218 cases of NSCLC, McMillen et al. correlated EGFR expression status with mutation status.[36] Phosphorylation at Y1045 was noted in 52% of cases, 71% of which exhibited the presence of an EGFR mutation. Phosphorylation of Y1068 was seen in 55% of cases, but it was present in 73% of those cases with an EGFR mutation. The data demonstrate that among Chinese patients, immunohistochemical detection of p-1045 and p-1068 expression predicts EGFR mutations.

Activation mutations identified within the kinase domain of the EGFR gene led researchers to propose and develop therapeutic strategies targeting EGFR TK. Therapeutic strategies targeting the EGFR pathway offered exciting new options for the treatment of NSCLC. EGFR alterations have prompted the development of two classes of anti-EGFR agents: monoclonal anti-EGFR antibodies (e.g., cetuximab and panitumumab) and small-molecule TKIs of EGFR (e.g., gefitinib and erlotinib, among others). According to large-cohort Phase III clinical trials,[37–40] the response rates range from 15 to 37.5%. Clinical trials were initiated that employed novel agents targeting the EGFR TK. The results of these clinical trials indicated that many of the tumors harboring mutant EGFR are highly sensitive to EGFR TKIs, with 10–30% demonstrating a significant clinical response.[15,41]

Summary

Constitutive activation of EGFR TK in NSCLC is associated with carcinogenesis of NSCLC.

The incidence of EGFR mutations in NSCLC is dependent upon gender, smoking history, tumor type and ethnic background.

Female gender, zero or very light smoking history, East Asian ethnicity, adenocarcinoma or bronchioloalveolar histology correlates with objective responses to single-agent TKI therapy in NSCLC.

EGFR Pathway Alteration & Implications

EGFR, located at chromosome 7p12, spans approximately 200 kb and contains 28 exons. It is a member of the ErbB family of four closely related TK receptors: EGFR (ErbB1), HER2/c-neu (ErbB2), HER3 (ErbB3) and HER4 (ErbB4).[42,43] EGFR is activated by binding of its specific ligands. Structurally, EGFR is composed of an N-terminal extracellular ligand-binding domain, a transmembrane lipophilic segment, and a C-terminal intracellular region containing a TK domain. Multiple ligands that bind and activate EGFR have been described, including EGF and TGF-α. Upon ligand binding to EGFR, the receptors form homo- or hetero-dimers, which activate their intrinsic intracellular protein TK. The ligand binding-induced dimerization results in cross-autophosphorylation of key tyrosine residues in the cytoplasmic domain, which function as docking sites for downstream signal transducers.[44] This activation of EGFR results in initiation of signaling cascades involving several downstream pathways.[4,26,45–50] Through its influence on these pathways, EGFR induces a number of crucial cellular responses, such as proliferation, differentiation, motility and enhanced cell survival (Figure 2).[50–53]

CaptureFigure2.PNG
Figure 2.

EGFR and its signaling pathway. Structurally, an EGF receptor (EGFR) monomer is composed of an extracellular domain consisting of two ligand-binding subdomains: a transmembrane lipophilic segment and an intracellular region containing tyrosine kinase domains that occupy exons 18–24. The binding of ligands to EGFR results in autophosphorylation of key tyrosine residues in the cytoplasmic domain and activation of its intrinsic, intracellular protein tyrosine kinase activity. EGFR activation results in initiation of signaling cascades. These function to further modulate cell proliferation and survival through two downstream intermediate pathways: the PI3K–AKT–mTOR pathway and the RAS–RAF–MEK–MAPK pathway. These two intermediate pathways influence several key aspects of the cell cycle that include cell proliferation, apoptosis, migration and survival, and more complex processes such as angiogenesis.

P: Phosphorylation.

One of the major molecular alterations in the carcinogenesis of NSCLC is the activation mutation of EGFR. The mechanisms that regulate EGFR expression, such as epigenetic alteration and aberrant transcription factors, have been studied, but with inconclusive results. The significance of miRNA-128b was reported by Weiss et al., who found loss of miRNA-128b in two out of three NSCLC cell lines and in tumors from 55% of NSCLC patients.[54] miRNA-128b directly regulated EGFR expression, and loss of miRNA-128b correlated with better tumor responsiveness to gefitinib treatment and improved survival (23.4 vs 10.5 months).[54]

A strong genetic association with particular germline mutations has been shown to influence the susceptibility to EGFR TKIs (i.e., those that confer mutations in EGFR signaling). Liu et al. found that the frequencies of the -216T and CA-19 alleles are significantly higher in patients with any mutation, in particular in those with exon 19 microdeletions.[55] The -216T allele is preferentially amplified in human lung cancer specimens and cancer cell lines. These results suggest that the local haplotype structures across the EGFR gene may favor the development of carcinogenesis and thus significantly confer risk to the occurrence of EGFR mutations in NSCLC, particularly the exon 19-microdeleted cases.[55] A novel germline transmission of the EGFR mutation V843I in a family with multiple members with lung cancer has been reported.[56] The proband was a 70-year-old woman who had multiple adenocarcinomas with EGFR mutations. These observations suggest that germline EGFR V843I mutation may result in altered EGFR signaling in cases of multicentric adenocarcinoma, bronchioloalveolar carcinoma and atypical adenomatous hyperplasia, and may also play a role in the development of lung cancer in multiple family members.[56]

EGFR plays a key role in the growth and survival of many solid tumor types.[21,57] Mutations affecting EGFR activity or expression can result in cancer.[58] The EGFR TK modulates cell proliferation and survival through two downstream intermediate pathways: the PI3K–AKT–mTOR pathway and the RAS–RAF–MEK–MAPK pathway.[59] These downstream cell signaling pathways influence several critical cellular processes, including cell proliferation, apoptosis, migration and survival. They are also involved in more complex processes such as angiogenesis and tumorigenesis. Studies on EGFR oncogene activation have been focused on gene mutations, DNA copy number alterations, protein expression alterations and genetic alterations of downstream signaling molecules.

Results from a Phase III trial evaluating the EGFR TKI, gefitinib, indicated that approximately 10% of patients responded to the therapy, and no survival benefit was observed.[30] Follow-up analysis identified mutations in the TK domain of EGFR in eight out of nine responders, whereas no mutations were detectable in seven patients who did not respond to gefitinib therapy.[12]

The most widely used EGFR TKIs are gefitinib and erlotinib. These agents are reversible inhibitors that compete with ATP at the active site of the TK receptor domain.[6] They are primarily used in patients who have failed platinum-based chemotherapy.[60] In a randomized Phase III study, erlotinib significantly improved median survival from 4.7 (placebo) to 6.7 months in patients with NSCLC who had previously failed one or two chemotherapy regimens.[61] Gefitinib improved disease-related symptoms in heavily pretreated symptomatic patients with NSCLC.[30] However, in the Phase III Iressa Survival Evaluation in Advanced Lung Cancer (ISEL) trial, which included pretreated patients with recurrent disease, gefitinib failed to demonstrate a survival benefit in the overall unselected patient population compared with placebo-treated controls.[62] Recently, however, results from the randomized Phase III IRESSA NSCLC Trial Evaluating Response and Survival against Taxotere (INTEREST) indicate that gefitinib is not inferior to docetaxel in terms of overall survival, suggesting that this TKI may also be a viable option for previously treated patients with advanced NSCLC.[63]

The data from 222 publications indicate that EGFR mutations are predictive of patient response to single-agent EGFR TKI treatment in advanced NSCLC.[64]

EGFR Mutations

The EGFR mutations responsible for the constitutive activation of receptor TK are also most frequently associated with sensitivity to EGFR TKIs.[65] These mutations are associated with response rates of >70% in patients treated with either erlotinib or gefitinib.[28,66]

Receptors containing different mutations appear to have different signaling properties, but most mutations seem to affect the ATP-binding cleft, which is also where targeting TKIs bind (Figure 3).[49]

CaptureFigure3.PNG
Figure 3.

Mechanism of EGFR-activating mutations and EGFR-targeted therapy. (A) EGFR mutations render EGFR tyrosine kinase constitutively activated. Activated EGFR phosphorylates key tyrosine residues (P) in the tyrosine domain, which initiates downstream effectors. (B) At present, two categories of agents are used for inhibiting EGFR signaling: humanized antibodies and small-molecule TKIs. Antibodies inhibit ligand-dependent activation of EGFR by blocking the ligand-binding site and preventing activation. TKIs block the magnesium–ATP-binding pocket of the intracellular tyrosine kinase domain, further inhibiting autophosphorylation. This inhibition disrupts tyrosine kinase activity and abrogates intracellular downstream signaling.

TKI: Tyrosine kinase inhibitor.

In vitro studies have demonstrated that mutant EGFR has enhanced TK activity, leading to a greater sensitivity to anti-EGFR inhibition. As mentioned previously, the four most common mutations seem to be those most closely associated with TKI sensitivity. The discovery that many objective responders to TKIs harbored EGFR mutations in exons 19 and 21 was a major breakthrough in patient selection for EGFR targeting therapy.[67,68] The most frequent mutation, located in exon 19, eliminates four amino acids – leucine, arginine, glutamic acid and alanine – downstream from the lysine residue at position 745.[67,69–72] Patients with an EGFR mutation who were treated with TKI had much higher response rates and longer progression-free survival than those without a mutation (Table 1).[69]

Both retrospective and prospective studies have demonstrated that NSCLC patients carrying the described EGFR gene mutations have a significantly higher response rate to gefitinib and/or erlotinib compared with patients with wild-type EGFR.[12,15,16,28,67,68,73–76] Some patients experienced rapid complete or partial responses that were durable.[26] The discovery of somatic mutations in EGFR that correlated with sensitivity to TKIs identified a plausible and reproducible explanation for these observations.

The most commonly used methods to detect mutations are direct sequencing and real-time PCR.[73,77] Other methods include single-strand conformational polymorphism analysis[78,79] and high-resolution melting amplicon analysis.[17,80] Scorpion ARMS® (QIAGEN, Germany), a multitargeted real-time PCR detection kit, allows the detection of the most prevalent somatic mutations in the EGFR that are common in human cancers. The high sensitivity and specificity of the kit permits the detection of mutations against a background of wild-type genomic DNA. The kit uses DxS Scorpions® (QIAGEN) technology to detect exon 19 deletions and mutations in exons 19–21 (T790M, L858R, L861Q, G719X and S768I) and any one of three insertions into exon 19 (2307_2308ins9, 2319_2320insCAC and 2310_2311insGGT). Relative to the direct sequencing method, the other two techniques allow for the rapid detection of EGFR mutations with high sensitivity and specificity. However, confirmation of mutations via direct sequencing is necessary.[24,80,81] Standardization is essential for the clinical application of EGFR mutation tests. However, at present, there is no official guideline for these EGFR mutation tests. The number of mutation sites that are needed in the testing protocol still remains to be established. Large-scale clinical trials are also needed.

Jackman et al. studied 223 chemotherapy-naive patients with advanced NSCLC.[28] Sensitizing EGFR mutations were associated with a 67% response rate, with a time to progression (TTP) of 11.8 months and overall survival of 23.9 months. Exon 19 deletions were associated with a longer median TTP and overall survival compared with L858R (exon 21) mutations. Wild-type EGFR was associated with poor outcomes (response rate: 3%; TTP: 3.2 months), irrespective of KRAS status. EGFR genotype was more effective than clinical characteristics at selecting appropriate patients for consideration of first-line therapy with an EGFR TKI.

Studies indicate that more than 75% of patients responsive to TKI therapy have activating mutations in EGFR.[13,77] However, some rare types of EGFR mutations can confer resistance to EGFR-targeted therapies after treatment with TKIs when combined with the common activating mutations.[82–85] Clinically, patients with EGFR exon 20 mutations do not respond to gefitinib.[72] Moreover, the appearance of a secondary mutation in exon 20 (T790M) accounts for approximately 50% of acquired drug resistance.[77,86] Screening for the emergence of such mutations in circulating tumor cells from the blood of patients during the course of treatment may allow earlier identification of acquired resistance.[83,87]

Results of some preclinical studies suggest that the clinical benefit observed with EGFR TKIs is not restricted to those patients harboring EGFR mutations. This may be due to molecular factors outside of gene mutations. EGFR amplification and receptor/ligand overexpression, both of which allow for a 'gain of function' to occur, are implicated in creating a scenario of EGFR dependence that causes the sensitivity to single-agent EGFR inhibitors.[44,88] However, the data from the IRESSA Pan-Asia Study (IPASS) clearly demonstrate that patients with increased EGFR copy numbers and no EGFR mutations do not benefit from EGFR TKIs.[89]

EGFR Copy Number Alterations

EGFR is frequently over-represented or amplified in NSCLC, which is commonly associated with EGFR overexpression.[90] Increased EGFR copy numbers may result from gene amplification or polysomy of chromosome 7. The incidence of EGFR amplification ranges from 12 to 59%, depending on the patients selected and the technology used.[64,88,91,92] Some, but not all, studies have revealed that positive EGFR amplification is associated with significantly better survival after treatment with a TKI.[88,93] Gain of EGFR copy number has been consistently associated with a favorable outcome after EGFR TKI therapy; it has also been proposed to be a potential biomarker of TKI responsiveness.[91,94]

Somatic EGFR mutations consistently correlate with improved response rates; by contrast, the results of studies investigating EGFR copy number as a predictor of response to TKIs have been inconsistent.[25,95] Overall, EGFR mutations seem to have higher sensitivity and specificity for predicting response to TKIs than EGFR copy number gain status.[64] High copy numbers of EGFR have been detected in approximately 30% of NSCLC patients using FISH, and are reportedly associated with better responses to TKI therapy,[73,96] although the EGFR mutation status of those cases was unclear. Approximately 70% of patients with EGFR copy number gain also had EGFR somatic mutations, a fact that clouds the true significance of EGFR copy number gain. IPASS demonstrated that EGFR mutation was the strongest predictor of improved progression-free survival. There was a high degree of overlap between EGFR mutation positivity and high EGFR gene copy numbers: of 245 patients with high EGFR copy numbers whose EGFR mutation status was also known, 190 (78%) were also EGFR mutation-positive. This suggests that the improved outcome in high EGFR copy number patients is being driven by the EGFR mutation-positive overlap.[89] Some researchers have suggested that high EGFR copy numbers can be used as a predictive biomarker for response and survival benefit in patients with NSCLC who receive EGFR TKI therapy.[88] However, the data from the IPASS trial clearly demonstrate that patients with increased EGFR copy numbers and no EGFR mutations do not benefit from EGFR TKIs.[89] Hirsch et al. suggested that although EGFR mutations and high copy numbers are both predictive of response to erlotinib in NSCLC, EGFR copy number was a more powerful predictor of differential survival benefit from erlotinib.[88]

There are several methods for detecting and determining EGFR copy number and dosage, including FISH,[73,88,97] chromogenic in situ hybridization[92,98] and real-time quantitative PCR.[20,99,100] When EGFR copy number was measured by PCR, it was found that increased EGFR copy number was significantly associated with prolonged survival, indicating a potential prognostic value of EGFR copy number.[64,101] The patients with EGFR gain demonstrated a higher disease control rate (67 vs 26%), longer TTP (9.0 vs 2.5 months) and prolonged survival time (18.7 vs 7.0 months).[64] It is noteworthy that EGFR copy number is used as a predictor for response to TKI therapy largely because it is correlated with EGFR mutations – EGFR mutations are the best predictors. Hirsch et al. investigated 229 chemotherapy-naive patients with advanced-stage NSCLC in a Phase II clinical trial.[88] Among the 76 patients analyzed by FISH, 59.2% had increased EGFR copy numbers, as indicated by four or more gene copies per cell in >40% of the cells. Response was higher in EGFR-amplified patients (45%) versus the EGFR-unamplified patients (26%). Those patients with EGFR amplification had a median progression-free survival time of 6 months compared with 3 months for patients without amplification. Median overall survival was 15 months for the EGFR-amplified group, while it was only 7 months for patients without amplification.[102]

Despite a majority of studies demonstrating that high EGFR copy number correlates with better response and increased survival in NSCLC patients treated with EGFR TKIs, debate remains about its true predictive value. Some studies suggest that when compared with EGFR mutations, EGFR gene copy number is a less sensitive and less specific marker and may not be considered clinically suitable for patient selection.[64] Douillard et al. also reported that EGFR mutation demonstrates greater predictive power than EGFR copy number in therapy response and progression-free survival.[103] Further studies are necessary to resolve these discrepant findings.

EGFR Overexpression

The clinical implications of EGFR overexpression have been studied extensively. However, the results have been inconclusive thus far. Immunohistochemistry-based assays measuring EGFR expression could not reliably predict the response to EGFR TKI therapy. Overexpression of EGFR has been demonstrated in 40–80% of cases of NSCLC and has been associated with a poor prognosis.[104–106] The initial assumption was that EGFR antibodies would be more effective in tumors with robust overexpression of EGFR. However, early clinical studies were unable to demonstrate a distinct correlation between EGFR expression and the likelihood of response to EGFR inhibition with targeted antibodies.[107] In addition, studies suggest that immunohistochemistry-based assays measuring EGFR expression do not serve as reliable predictors of response to cetuximab therapy.[108]

Increased response rates after treatment with a TKI have been demonstrated in patients with positive EGFR immunostaining in some studies, but not in others.[104,105,109,110] In multivariate analyses, EGFR expression level was associated with an objective response or adverse prognosis in NSCLC.[93,110] Several investigations into the prognostic significance of EGFR expression revealed no association with survival benefit.[102,105,111] Therefore, EGFR overexpression by itself is not prognostic of survival in NSCLC. It has been suggested that the nonoptimized cut-off value for EGFR-positive immunostaining and/or lack of standardization in staining procedures and guidelines may explain the discordance among these studies.[111]

EGFR Mutation-specific Antibodies

Since the use of EGFR overexpression as a prognostic marker has largely been unproductive, considerable efforts have been made to develop antibodies that react specifically with the mutant form of EGFR. Cell Signaling Technology, Inc. (MA, USA) has developed two mutant-specific antibodies against the most common mutant forms of EGFR: the 15-base pair/5-amino acid deletion (E746-A750del) in exon 19 and the L858R point mutation in exon 21.[112] Yu et al. investigated EGFR genotypes of 40 NSCLC tumor samples by immunohistochemistry with these antibodies and confirmed the immunohistochemistry results by DNA sequencing.[112] Detection of mutant EGFR by these two antibodies was performed by western blotting, immunofluorescence and immunohistochemistry. The sensitivity and specificity of these antibodies in a 340-sample panel of paraffin-embedded NSCLC tumors was 92 and 99%, respectively, compared with direct sequencing and mass spectrometry-based DNA sequencing. These results demonstrate that mutation-specific antibodies provide a rapid, sensitive, specific and cost–effective method to identify lung cancer patients who may respond to EGFR-targeted therapies. Brevet et al. evaluated the two mutation-specific monoclonal antibodies for the detection of EGFR mutations by immunohistochemistry on 218 paraffin-embedded lung adenocarcinomas.[112] The EFGR L858R mutant antibody showed a sensitivity of 95%, a positive predictive value of 99% and a specificity of 76%, with a positive cut-off of (2+).[113] A positive threshold of (2+) will effectively reduce the false-positive rate and enhance the predictive power of immunohistochemistry assays to 100%, with a minimal reduction in sensitivity. The immunostaining scoring was based on cytoplasmic and/or membrane staining intensity as follows: (0+) = no staining or faint staining intensity in <10% of tumor cells; (1+) = faint staining in >10% of tumor cells; (2+) = moderate staining; and (3+) = strong staining. Therefore, immunohistochemistry using mutation-specific antibodies can be used to screen for patients who may be candidates for EGFR inhibitors.[113]

Phosphorylated Form of EGFR

Aberrant activation of EGFR is a recognized component of cancer development and progression.[59] In addition, recent data indicate that both EGFR mutations and the activation status of EGFR, defined by phosphorylation, might have a strong impact on the clinical course of NSCLC.[114] The two major EGFR signaling pathways, PI3K–AKT–mTOR and RAS–RAF–MAPK, mediate EGFR effects on cell proliferation and survival. The activation of these pathways is dependent on the phosphorylation status of the components. Investigations to date indicate that the major molecular alteration involved in the carcinogenesis of NSCLC is an activation mutation. The mechanisms that regulate EGFR expression, such as epigenetic alteration and aberrant transcription factors have been studied but are not yet conclusive.

Phosphorylation at tyrosine 845 in the kinase domain of EGFR may stabilize the activation loop, which maintains the receptor in an active state and provides a binding surface for substrate proteins.[115] Phosphorylation of two additional tyrosines, 1068 and 1173, mediates the direct binding of growth factor receptor-bound protein 2. Furthermore, tyrosine 1068 is involved in the activation of the MAPK signaling pathway.[116]

Detection of activated EGFR is conducted by using anti-phospho-EGFR antibodies directed at EGFR in its phosphorylated state. Phosphorylations in the carboxyl-terminus of EGFR play a key role in the recruitment of signaling molecules and activation of downstream signaling pathways.[115,117] In a study by Endoh et al., involving 97 NSCLC cases, patients with phospho-EGFR-positive tumors demonstrated a prolonged survival, although the follow-up period was relatively short.[117] Hijiya et al. investigated 21 cases of NSCLC for correlations between the presence of EGFR mutations and the EGFR phosphorylation status by immunohistochemistry with antibodies recognizing EGFR that was phosphorylated at tyrosine 992 and tyrosine 1173, respectively.[118] The mutation status of EGFR was strongly correlated with immunoreactivity for phosphorylated tyrosine 992, indicating a clear potential for using anti-phospho-EGFR antibodies as a surrogate marker of EGFR mutations and thus predicting the clinical response to tyrosine antagonist therapy.

The immunohistochemical evaluation of NSCLC with anti-phospho-EGFR antibodies is potentially useful in the clinical prediction of responsiveness to EGFR-targeted therapy. However, further testing and evaluation are needed to determine its true clinical implication.

EGFRvIII

The newly characterized EGFR mutant, EGFRvIII, results from an in-frame deletion of exons 2–7 of the coding sequence, which has been found to be generated by gene rearrangement or aberrant mRNA splicing.[119,120] The variant form has a deletion of 267 amino acids in the extracellular domain of normal EGFR, creating a unique epitope at the fusion junction. A number of functional differences between EGFRvIII and EGFR have been characterized.[120,121] Although EGFRvIII fails to bind EGF, its intracellular TK is constitutively activated, allowing the receptor to undergo tyrosine autophosphorylation.[122,123] These studies provide further evidence that EGFRvIII expressed in NSCLC is phosphorylated and, hence, activated. The data suggest that the sustained activation of EGFRvIII may play a role in the pathogenesis of NSCLC and, therefore, EGFRvIII is a potential therapeutic target for NSCLC.[114] Antibodies directed to this tumor-specific variant of EGFR provide an alternative targeting strategy. It has been demonstrated that systemic treatment of mice bearing tumors expressing EGFRvIII with monoclonal antibodies specific for EGFRvIII inhibited tumor growth and extended animal survival.[124,125] Antibodies that have an affinity for EGFRvIII, but without an affinity to wild-type EGFR, provide an alternative tool for detecting this mutation variant.[126]

The role of EGFRvIII mutations in the pathogenesis of NSCLC is unclear. Reported incidences of EGFRvIII mutation in NSCLC vary from 0 to 42%.[127,128] These differences may be due to differences in the tumor composition (histological type) or to technical considerations, such as the threshold for the result interpretations. Studies using immunohistochemical assays with EGFRvIII mutant-specific antibodies suggest that this mutation is present in multiple other tumor types and is not exclusive to NSCLC.[114,129] However, owing to the large size and complex genomic structure (28 exons spanning ~190 kb) of EGFR and its large intron 1 (123 kb), where genomic deletions frequently occur, it has been difficult to assess and verify the existence of the EGFRvIII mutations at the genomic level.[127] To evaluate the clinical impact of EGFRvIII in NSCLC, Okamoto et al. investigated EGFRvIII expression in 76 cases of NSCLC by immunohistochemistry, using a monoclonal antibody specific for this EGFR variant. EGFRvIII expression was found in 39% (30/76) of NSCLC; however, genetic analysis of EGFRvIII mutations only generated a 3% positive rate compared with the 39% immunopositivity rate.[114] Okamoto et al. found that EGFRvIII was also observed in several normal tissue components of the lung, which raised the question of the clinical implications for this detection methodology.[114]

Studies of small-molecule TKIs have demonstrated clinical responses in NSCLC patients whose tumors bear EGFR kinase domain mutations. However, the efficacy of these inhibitors against tumors with the EGFRvIII mutation remains unclear. Ji et al. determined that EGFRvIII mutations were present in 5% (3/56) of human squamous cell lung carcinomas, but found no EGFRvIII mutations in a large cohort of human lung adenocarcinomas (0/123).[127] In their study, EGFRvIII-bearing tumors seemed relatively resistant to some TKIs, but responsive to others.[130] In an in vivo system, treatment with an irreversible EGFR inhibitor, HKI-272, dramatically reduced the size of EGFRvIII-driven murine tumors within 1 week.[126] A total of 7 days of erlotinib treatment led to an average reduction of 45% in tumor volume in the three treated mice. By contrast, those treated with HKI-272 demonstrated a reduction of 88%. The Ba/F3 cells, transformed with the EGFRvIII mutant, were relatively resistant to gefitinib and erlotinib in vitro, but sensitive to HKI-272, suggesting that TKI treatment is potentially efficacious for cancers harboring the EGFRvIII mutation.

Summary

Current studies of alterations of the EGFR pathway have been focused on gene mutations, gene copy-number alterations, protein expression alterations and downstream genetic alterations.

Four activating mutations – exon 18 (G719A/C), exon 21 (L858R and L861Q), and in-frame deletions in exon 19 – are the dominant mutations present in NSCLCs.

Patients with an EGFR mutation, who were treated with TKIs, had much higher response rates and longer progression-free survival than patients without EGFR mutations who had the same treatment.

Acquisition of a new mutation in exon 20 can confer resistance to TKI treatment.

Overexpression of EGFR has been found in 40–80% of cases, but its usefulness as a predictive marker remains controversial.

Sustained activation of EGFRvIII is implicated in the pathogenesis of squamous cell carcinoma and, thus, EGFRvIII is a potential therapeutic target in this subset of NSCLCs.


EGFR-targeted Therapy & Mechanisms of Resistance

There are two major approaches for inhibiting EGFR signaling: to prevent ligand binding to the extracellular domain with a monoclonal antibody and to inhibit the intracellular TK activity with a small molecule. Use of the latter approach was the first method to be attempted clinically.[131] The EGFR TKIs are reversible competitive inhibitors of the TK domain of EGFR that bind to its ATP-binding site. Somatic activating mutations of the EGFR gene, increased gene copy number and certain clinical and pathological features have been associated with dramatic tumor responses and favorable clinical outcomes with these agents in patients with NSCLC. The majority of these patients inevitably acquire resistance to EGFR TKIs. Recent data indicate that a secondary mutation, such as T790M, expression of HGF, PTEN and/or early growth response-1 and changes in the epithelial-to-mesenchymal transition, were associated with EGFR TKI resistance. Uramoto et al. found that strong expression of HGF was detected in six out of eight specimens with the T790M mutation.[132] Three out of eight cases (38%) demonstrated a loss of PTEN in samples with the T790M mutation. A loss of early growth response-1 was detected in two out of seven cases (29%), including one tumor without PTEN. Four out of seven cases (57%) demonstrated positive expression of phosphorylated Akt. A change in the epithelial-to-mesenchymal transition status between pre- and post-treatment was observed in four out of nine cases (44%). These results suggest that alterations in gene or protein expression can account for all mechanisms by which tumors acquire resistance to EGFR TKIs.[132] This phenomenon suggests the existence of complicated relationships between acquired resistance-related genes.

Somatic activating mutations in EGFR are identified in a subset of NSCLC that responds to TKIs. As noted previously, acquisition of drug resistance has been linked to a specific secondary somatic mutation, EGFR T790M. Bell et al. described a family in which multiple members developed NSCLC associated with germline EGFR mutations of T790M.[25] These observations implicate altered EGFR signaling as a culprit in the genetic susceptibility to lung cancer in families with an increased incidence of NSCLC.

We propose an algorithm for molecular testing for patients with NSCLC (Figure 4). A stepwise approach, based on the frequency of specific mutations, is used to assess lung cancer patients for specific findings, which will allow proper therapeutic stratification for targeted therapy.


Figure 4.

Suggested algorithm for molecular testing for patients with non-small-cell lung cancer. A stepwise approach is used to test lung cancer patients according to the known frequencies of various mutations. Small-cell lung cancers are excluded from testing. NSCLC, which accounts for approximately 85% of all lung cancers, is tested for the presence of EGFR mutations. A positive test, found in approximately 20% of Caucasians and 40% of East Asians, predicts an 80% probability of response to EGFR TKI therapy. Nonmutated EGFR is found in approximately 80% of Caucasians and 60% of East Asians. These patients are further tested for EML4–ALK mutations. EML4–ALK mutations are found in only 3% of patients with NSCLC, but the mutation predicts a 53% probability of response to targeted therapy. Cases lacking EML4–ALK mutations may undergo additional testing.

EGFR: EGF receptor; Mu: Mutation; NSCLC: Non-small-cell lung cancer; SCC: Small-cell carcinoma; TKI: Tyrosine kinase inhibitor.

EGFR-targeted Therapy Approaches

Monoclonal Antibodies Monoclonal antibodies, such as cetuximab and panitumumab, are either chimeric mouse–human or fully humanized antibodies targeting the extracellular domain of EGFR and thereby inhibiting the binding of activating ligands to the receptor. This class of treatment inhibits ligand-dependent activation of EGFR and inhibits the downstream pathways, which cause cell cycle progression, cell growth and angiogenesis (Figure 3A). In addition, binding of the antibody initiates EGFR internalization and degradation, which leads to signal termination.[108,133] Fully humanized antibodies such as panitumumab, have a high affinity for EGFR and a longer half-life.[134] Although EGFR is frequently expressed in patients with NSCLC, the clinical efficacy of treatment with anti-EGFR antibodies is limited to only a subset of patients.

Tyrosine kinase inhibitors Tyrosine kinase inhibitors are synthetic small molecules that block the magnesium–ATP-binding pocket of the intracellular TK domain.[108] Several TKIs, such as gefitinib and erlotinib, are specific for EGFR, whereas others inhibit other receptors in addition to EGFR, such as HER2 and VEGF receptor 2. TKIs block ligand-induced receptor autophosphorylation by binding to the TK domain and disrupting TK activity, thereby abrogating intracellular downstream signaling (Figure 3B & Table 1).

Mechanisms of Resistance to EGFR-targeted Therapy

Acquired Resistance Caused by a Secondary Mutation Although EGFR mutations are associated with enhanced sensitivity to gefitinib and erlotinib, not all tumors that have activating mutations are associated with an enhanced response. The efficacy of EGFR TKIs is limited owing to either primary or acquired resistance after therapy. Most patients who initially respond to gefitinib and erlotinib eventually become resistant and experience progressive disease.

It is known that four mutations result in TKI drug sensitivity: point mutations in exon 18 (G719A/C) and exon 21 (L858R and L861Q), as well as in-frame deletions in exon 19, which eliminate four amino acids – leucine, arginine, glutamic acid and alanine – downstream of the lysine residue at position 745.[67,69–72] However, insertion mutations of exon 20 at D770–N771 were associated with EGFR TKI resistance.[60,135] This observation was confirmed in an in vitro model in which insertion mutations in exon 20 rendered transformed cells less responsive to EGFR TKIs compared with the sensitizing mutations of exons 19 and 21.[135]

Two established mechanisms of acquired resistance consist of additional mutations in the EGFR gene acquired during the course of treatment that change the protein coding sequence and amplification of other oncogene signaling pathways.[85,136–138]

Kobayashi et al. reported a gefitinib-resistant advanced NSCLC patient who had a relapse after 2 years of complete remission due to treatment with gefitinib.[86] The DNA sequence of EGFR at relapse revealed the presence of a second point mutation, resulting in a T790M mutation of EGFR. Structural modeling and biochemical studies showed that this second mutation led to gefitinib resistance.[86] The same mutation was confirmed by Pao et al. through molecular analysis of EGFR in patients with acquired resistance to gefitinib or erlotinib.[139] The gefitinib-resistant cases contain the same secondary mutation (T790M) in the kinase domain.[22] Codon 790 of EGFR is considered to be the 'gatekeeper' residue, which is an important determinant of inhibitor specificity in the ATP-binding pocket of EGFR.[108] Substitution of this residue in EGFR with a bulky methionine may cause resistance by steric interference with the binding of TKIs, including gefitinib and erlotinib.[86,139,140] This mutation may confer a survival advantage to the tumor and is probably selected for while the patient is receiving anti-EGFR TKI treatment.[25,84] These findings have led to the development of irreversible EGFR TKIs in an effort to effectively target this mechanism of resistance.[140]

The role of oncogenic activation of EGFR downstream effectors, such as KRAS, BRAF, PIK3CA and PTEN, in response to therapy is discussed extensively in a series of studies.[47,53] The RAS–MAPK and PI3K–AKT pathways are major signaling networks linking EGFR activation to cell proliferation and survival.[141,142] Mutations in these downstream effectors of EGFR signaling could lead to resistance to EGFR inhibitors.[136–138] The discovery of molecular aberrations, such as MET kinase amplification or mutations of EML4–ALK fusion, which causes constitutive activation of RAS–RAF–MEK, has provided further insight and validation into factors limiting the therapeutic efficacy of EGFR inhibitors.[46,143,144]

KRAS Mutations KRAS plays a key role in the EGFR signaling network. The KRAS proto-oncogene encodes KRAS G-protein, which plays a critical role in the RAS–MAPK signaling pathway downstream of many growth factor receptors, including EGFR.

One of the most important discoveries for the clinical management of colorectal carcinoma has been the association of mutations in KRAS and the efficacy of monoclonal antibodies targeting EGFR, such as panitumumab and cetuximab. Some tumors harbor somatic mutations in exon 2 of KRAS that compromise the hydrolysis of RAS-bound GTP to GDP, resulting in constitutive activation of the RAS pathway.[145] In the presence of a KRAS mutation, EGFR pathway activation cannot be significantly inhibited by cetuximab or panitumumab, which acts upstream of the KRAS protein, diminishing the efficacy of the agents.

An activating mutation of KRAS is present in 15–30% of NSCLC cases[26,146,147] and accounts for approximately 35–45% of TKI-nonresponsive cases.[148] Approximately 30% of lung adenocarcinomas contain activating KRAS mutations, which are associated with resistance to EGFR TKIs.[22] It is noteworthy that the presence of a KRAS mutation is common in NSCLC, but the occurrences of KRAS and EGFR mutations seem to be mutually exclusive.[4,27,149–152] EGFR and KRAS mutations are rarely if ever detected in the same tumor, suggesting that they may perform functionally equivalent roles in lung tumorigenesis.[58,153] However, there is growing evidence that coexistence of EGFR and KRAS mutations is possible,[27,151,154] although the frequency is low. Due to the limited number of cases, it is difficult to obtain conclusive results; however, the available data suggests a negative association between EGFR/KRAS mutation and EGFR TKI responsiveness.[27,151,154]

It remains unclear whether assessment of KRAS mutation status will prove to be clinically useful with regard to anti-EGFR therapy.[50] Although an association between the presence of a KRAS mutation and lack of response to EGFR TKIs has been observed, it remains indeterminate whether this association is clinically relevant with respect to progression-free and overall survival. Investigations of KRAS mutation status as a negative predictor of outcome after anti-EGFR therapy have been undertaken, but small sample sizes due to low prevalence of KRAS mutations have limited the power of such studies. Some investigators have reported that KRAS mutation is a negative predictor of response to anti-EGFR monoclonal antibodies and also an important mechanism of resistance to TKIs in NSCLC.[26] Unlike colorectal cancer, KRAS mutations do not seem to identify patients who do not benefit from anti-EGFR monoclonal antibodies in NSCLC.

KRAS mutations are almost exclusively detected in codons 12 and 13 of exon 2, which may result in EGFR-independent intracellular signal transduction activation. In a study by Eberhard et al., EGFR exons 18–21 and KRAS exon 2 mutations were investigated via sequencing in tumors of 274 patients.[27] KRAS mutations were present in 21% of tumors, which were associated with significantly decreased TTP and survival in patients treated with erlotinib plus chemotherapy. Others have reported that KRAS mutation status did not impact EGFR TKI therapy.[28] In a study that included 223 chemotherapy-naive patients with advanced NSCLC treated with erlotinib or gefitinib monotherapy, EGFR mutations were associated with a 67% response rate. Wild-type EGFR was associated with poorer outcomes, regardless of KRAS mutation status.[28]

A study by Wang et al. utilizing PCR-restriction fragment length polymorphism analysis investigated the KRAS mutations in codons 12 and 13 in 273 NSCLC cases.[155] Of the 120 patients who received EGFR TKI therapy, only 5.3% (one out of 19) of the patients with a KRAS mutation demonstrated a response compared with a 29.7% response rate for patients lacking a KRAS mutation.[152] Furthermore, the median progression-free survival time of patients with a KRAS mutation was 2.5 months compared with 8.8 months for patients with wild-type KRAS.

A meta-analysis by Linardou et al. provided empirical evidence that somatic mutations of the KRAS oncogene are highly specific negative predictors of response to single-agent EGFR TKIs in advanced NSCLC.[148] Among 17 publications, 165 out of 1008 (16%) NSCLC patients presented with KRAS mutations. The presence of KRAS mutations was significantly associated with an absence of response to TKIs in these patients.

Having an intact KRAS is necessary, but not sufficient, to derive benefit from EGFR inhibition, and additional mechanisms of resistance to EGFR inhibitors exist.

KRAS testing scenarios in the management of NSCLC are summarized in Figure 5. The incidence of KRAS mutations in NSCLC is reportedly up to 20%.[156] In the subset of tumors with KRAS mutations, less than 3% also contain an EGFR mutation, and the remaining 97% of tumors with KRAS mutations have wild-type EGFR. Both KRAS/EGFR double mutations and wild-type EGFR are associated with nonresponsiveness to EGFR-targeted therapy.

Figure 5.


KRAS testing scenarios in the management of non-small-cell lung cancer. NSCLC accounts for approximately 85% of all lung cancers. The incidence of KRAS mutations in NSCLC is reportedly up to 20%. In the subset of patients with KRAS mutation, less than 3% of the tumors also contain an EGFR mutation; the remaining 97% have wild-type EGFR. Both KRAS/EGFR double mutations and wild-type EGFR are associated with nonresponsiveness to EGFR-targeted therapy. Of the 80% of NSCLCs that do not have KRAS mutations, approximately 20% harbor EGFR mutations, which are associated with an 80% likelihood of response to EGFR TKI therapy.

EGFR: EGF receptor; Mu: Mutation; NSCLC: Non-small-cell lung cancer; TKI: Tyrosine kinase inhibitor.

lung without KRAS and EGFR mutations.[160] Of these, ten adenocarcinomas with the BRAF-V600E mutation were identified. BRAF mutations were reported more frequently in micropapillary lung adenocarcinoma.[161] De Oliveira Duarte Achcar and colleagues analyzed the clinical and molecular profile of 15 primary micropapillary adenocarcinomas and found BRAF mutations in three cases (20%). The BRAF-V600E mutation-bearing tumors had a slight female predilection (6:4 female:male). The elderly patients were found to have a greater than expected incidence of intralobar satellite nodules and N2 node involvement.[161] The adenocarcinomas were largely of mixed type, with a high incidence of papillary (80%) and lepidic growth (50%). However, due to the relatively small sample size, it is yet to be determined whether BRAF mutant tumors represent a distinct subset of lung adenocarcinoma.[162]

Mutations in BRAF have been shown to impair responsiveness to panitumumab or cetuximab in patients with colorectal carcinomas. This initial retrospective work was performed on a cohort of 132 patients.[163] The results showed that none of the patients who experienced a response displayed BRAF mutations, whereas 11 of 79 nonresponders (14.0%) carried a BRAF-V600E allele.

Nevertheless, BRAF mutations are rarely detected in NSCLC when compared with KRAS mutations.[162,164,165] A total of 80% of the reported mutations are located within the kinase domain of BRAF.[159] Brose et al. identified activating BRAF mutations in five out of 292 cases (1.7%) of NSCLC. Among these mutations, three were found in exon 11 and two in exon 15.[164] It has been proposed that mutations in BRAF, a downstream signaling molecule of EGFR, predict clinical response to EGFR inhibitors, but this has yet to be validated in a larger number of cases.[157] Notably, a single substitution of glutamic acid for valine at codon 600 (V600E) accounts for approximately 90% of the BRAF missense mutations found in human tumors.[159] Considering BRAF is a serine/threonine kinase that is commonly activated by a somatic point mutation in human cancer, it may provide new therapeutic opportunities in a subset of NSCLC.

ALK Rearrangement ALK encodes a TK receptor found in a number of fusion proteins consisting of the intracellular kinase domain of ALK and the amino terminal portions of different genes.[166,167] A subset of NSCLC cases harbor a transforming fusion gene, EML4–ALK, within the genome. To date, seven gene fusion variants have been reported in NSCLCs. All involve the intracellular TK domain of ALK starting at a portion encoded by exon 20.[168] This fusion is formed as the result of a small inversion within the short arm of chromosome 2 that joins EML4 to ALK, inv(2)(p21;p23), which encodes an activated TK protein.[169,170] Several other transforming EML4–ALK fusion gene variants have also been identified, involving various EML4 exons and ALK.[171,172] ALK can also fuse with some other rare fusion partners, such as KIF5B and TFG.[173]

Activated ALK is involved in the inhibition of apoptosis and the promotion of cellular proliferation through activation of downstream PI3K–AKT and MAPK signaling pathways.[174] The EML4–ALK fusion is a rare abnormality detected in approximately 6% of patients with NSCLC.[144,169,175] Fusion of the EML4–ALK gene and its associated EML4–ALK product may lead to constitutive activation of the RAS–RAF–MEK–MAPK pathway.[47] In addition, two other less frequent ALK fusions in lung cancer have been reported.[176]

Non-small-cell lung cancer cases harboring EML4–ALK are characteristically found to have wild-type EGFR, as well as wild-type KRAS.[177,178] In addition, patients with these tumors tend to be younger, have an advanced clinical stage at presentation, have never smoked and their tumors exhibit solid histology, often with a component of signet ring cells.[178,179]

Patients with this alteration demonstrated an extraordinary response to the MET–ALK inhibitor, PF-02341066, in a Phase I/II trial.[144] Ten out of 19 patients (53%) experienced objective responses. A total of 15 out of 19 patients (79%) demonstrated disease control at 8 weeks, lasting as long as 40 weeks in some. Only four patients demonstrated disease progression. Kwak et al. screened 1500 NSCLC patients and identified 82 patients with advanced ALK-positive disease.[180] The vast majority (96%) of these cancers were adenocarcinomas. A total of 94% of these patients had received at least one prior therapy. All of these patients were treated with crizotinib, an oral small-molecule ALK TKI. The disease control rate was 90%, which included a 57% response rate plus 33% with stable disease. Furthermore, Rodig et al. evaluated the incidence and the characteristics of ALK-rearranged lung adenocarcinomas within the western population to elucidate the optimal diagnostic modality to detect ALK rearrangements in routine clinical practice.[179] In their study, 358 lung adenocarcinomas were tested for ALK rearrangement by FISH and immunohistochemistry. ALK rearrangement again demonstrated an association with younger age, never having smoked, advanced clinical stage, and a solid histology with signet ring cells in some cases. Of note, none of the ALK-rearranged tumors harbored coexisting EGFR mutations. The results of this study demonstrate that ALK rearrangements are uncommon in the western population, but represent a distinct clinical entity with unique attributes and the possibility of distinct treatment options. For suspected cases, dual diagnostic testing with FISH and immunohistochemistry should be considered in order to accurately identify lung adenocarcinomas with ALK rearrangement.[179] A study by Zhang et al. involving 266 Chinese patients with NSCLC revealed approximately similar results.[178]

Shaw and colleagues investigated 141 NSCLC cases and found that 19 (13%) contained EML4–ALK fusion, 31 (22%) demonstrated EGFR mutantations and 91 (65%) were wild-type for both.[144] Several studies provide consistent evidence that EML4–ALK and EGFR mutations are mutually exclusive.[144,169,177,181] However, Tiseo et al. reported a 48-year-old Caucasian man who had never smoked and who was diagnosed with NSCLC with a concomitant EGFR mutation and ALK translocation that was resistant to erlotinib therapy.[182] This may be representative of the nature of this subgroup of NSCLC.[154]

No EGFR mutations in the EML4–ALK cohort and no instances of ALK rearrangement in the EGFR cohort have been found.[144] Among patients with metastatic disease, EML4–ALK rearrangements were associated with resistance to EGFR TKI treatment.

Compared with the EGFR-mutated and wild-type EGFR/ALK cohorts, patients with EML4–ALK fusion gene-positive tumors were significantly younger and more likely to be male.[144] However, another study demonstrated that males and females are equally affected.[139] Patients with EML4–ALK-positive tumors are more likely to be never/light smokers. A total of 18 of the 19 EML4–ALK-positive tumors were adenocarcinomas, predominantly of the signet ring cell subtype.[144] EML4–ALK defines a molecular subset of NSCLC with distinct clinical characteristics. Specifically, patients who harbor this mutation do not benefit from EGFR TKIs and should be directed to trials investigating ALK-targeted agents.

Summary

Two classes of anti-EGFR agents – monoclonal anti-EGFR antibodies and small-molecule EGFR TKIs – are currently used in EGFR-targeted therapy.

Additional mutations in EGFR are an important mechanism of acquired resistance to EGFR-targeted therapy.

Some mutations, such as exon 20 D770–N771 and T790M, are associated with EGFR TKI resistance.

Oncogenic activation of EGFR downstream effectors, such as KRAS, BRAF, PIK3CA and deletion of PTEN, is associated with resistance to TKI therapy.

KRAS mutation accounts for approximately 35–45% of TKI-nonresponsive NSCLC patients.

KRAS and BRAF mutations, as well as ALK rearrangements, are mutually exclusive with EGFR mutations.

ALK rearrangements are more common in younger patients who have never smoked, and their tumors often exhibit solid architecture, signet ring cell histology and wild-type EGFR and KRAS.

Conclusion

The EGFR is an effective therapeutic target in treating NSCLC. EGFR mutations have been used as a selection criterion for EGFR TKIs and are also used as a predictive marker for responsiveness to EGFR-targeted therapy. However, evidence is beginning to demonstrate that NSCLC may be composed of multiple subsets of tumors, each with its own molecular abnormalities.

Identification of the relevant molecular subtypes of this heterogeneous disease and selecting patients for the appropriate targeting agents is critical in the personalized therapy of NSCLC. KRAS/BRAF mutations, which are mutually exclusive with EGFR mutations, are rare in NSCLC but may be important mechanisms in the etiology and prediction of resistance to EGFR TKI therapy.

In conclusion, one unifying predictive model does not apply to all tumor types, and the larger goal of discovering a predictive marker to guide patient selection for EGFR-targeted therapy remains elusive. EGFR alteration markers need to be further evaluated in combination with clinical data to provide clear rationales for future therapeutic strategies in the treatment of NSCLC.


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-24 05:01:43 | 显示全部楼层 来自: 美国

往事不随风

本帖最后由 荷花池荒岛 于 2014-8-27 12:34 编辑


1)
zhqqiu82
五一期间,也给妈妈用了凡德他尼200mg/day,针对妈妈原本就多年的高血压,我把妈妈的降压药由原来的硝苯地平换成了厄贝沙坦氢氯噻嗪。对于凡德的心脏方面的不良反应是服用的辅酶Q10,剂量是200mg。准备了易蒙停,妈妈服药第一天晚上和次日早晨有轻微腹泻,后来正常。到5.8妈妈服用凡德第5天,妈妈说偶尔头疼,心脏无不适,无明显疲劳感。服药前两天,我和妈妈一起服用了凡德200mg/day,我没有服用辅酶Q10,在我身上凡德的副作用是头晕,轻微腹泻及乏力。但是心脏着实感受到了凡德的威力,时常心率变快,心悸的感觉也很明显,停药后能够立即好转。
http://www.yuaigongwu.com/forum. ... page%3D1&page=2   19楼


2)
乔乔
。。。。。。
母亲颈椎痛腰痛再次放疗以后,经历了白介素,特比奥,以及不断的血小板输液,血小板却一直升不上去,母亲身体一天比一天虚弱,从脚开始往上浮肿,声音嘶哑无法发声,我最爱的母亲于2013年1月20日病逝,循环系统及呼吸系统衰竭,去世之前的晚上一直高烧不退,喘不上来气,当母亲用尽最后力气对我说孩子放弃吧,我泪流满面,因为我知道母亲对于生命的渴望。。。
。。。。。。
http://www.yuaigongwu.com/thread-8910-1-13.html                   1楼


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:03:58 | 显示全部楼层 来自: 美国

化疗方案

本帖最后由 荷花池荒岛 于 2015-9-27 01:56 编辑

1)2013年非小细胞肺癌临床实践:改变了什么?
http://www.yuaigongwu.com/forum. ... amp;_dsign=edf7a7c0

2)晚期肺癌化疗及靶向药物治疗
http://wenku.baidu.com/view/b9ee21e5aeaad1f346933f4d.html

3)小剂量吉西他滨和多西他赛单药治疗老年晚期非小细胞肺癌
http://www.yuaigongwu.com/forum. ... &extra=page%3D4

4)肺癌常用的化疗方案
http://wenku.baidu.com/view/f4d328d549649b6648d7475e.html?re=view

常用肺癌化疗方案:
1、 对于非小细胞肺癌,紫杉醇+卡铂或顺铂,吉西他滨(健择)+卡铂或顺铂,多西他赛+卡铂或顺铂,培美曲赛+卡铂或顺铂。
2、小细胞肺癌:VP16+卡铂。  
3、顺铂效果比卡铂好,就是需要水化,相对麻烦些。
在以铂类为基础的化疗方案中,有些也应用第四代铂类产品,如奈达铂。


多西他赛化疗方案
  • 化疗前1天开始,口服0.75mg地塞米松6片每十二小时一次,持续至化疗结束后1天。
  • 化疗当天:多西他赛75mg/m2加入生理盐水500ml中(必须用玻璃瓶子,塑料瓶子可能会溶解),静脉点滴3小时以上,应用3B输液器或输血器;卡铂500毫克加入5%葡萄糖500毫升中,正常速度滴注。盐酸托烷司琼5毫克加入生理盐水100毫升中,化疗开始前,开始后静脉点滴各一次。
  • 第五天,复查血常规、肝肾功。  
  • 每周至少复查血常规2次,根据骨髓抑制程度注射升白药。可间断检查肝肾功能。血常规需查至化疗结束后至少两周。  
  • 其他副反应如便秘,脱发等可针对病情予以处理。盐酸托烷司琼可酌情换用其他抗呕吐药物。


长春瑞宾卡铂(NC)化疗方案
  • 第1天:化疗开始前,君凯9mg加入生理盐水100ml中静脉滴注。长春瑞宾40mg加入生理盐水50ml中备用。地塞米松5mg加入生理盐水100ml中,化疗前快速静脉滴入50ml,改换为长春瑞宾,10分钟内快速滴注。化疗药输完后立即快速滴注剩余的50ml地塞米松盐水。  
  • 第2天:化疗开始前,君凯9mg加入生理盐水100ml中静脉滴注。卡铂500mg加入5%葡萄糖溶液500ml中,静脉点滴4小时。  
  • 本周至少复查血常规2次。肝肾功能1次。
  • 第8天,同样方法静点长春瑞宾。  
  • 休息2周,中间每周至少复查血常规2次,根据骨髓抑制程度注射升白药。
  • 第22天,同样方法注射长春瑞宾。
  • 第23天,同样方法静脉点滴卡铂。
  • 第29天,同样方法静点长春瑞宾。 复查血常规及升白药用法同上。  
  • 长春瑞宾应用时注意勿使药物外渗,易造成皮肤坏死。  
  • 长春瑞宾约20%病人出现静脉炎,可对症处理,一般不至于出现严重后遗症。
  • 以上君凯等辅助用药可酌情换用其他类似药物。


紫杉醇卡铂(PC)化疗方案
  • 化疗前一天晚22点,化疗当日凌晨4点口服0.75mg地塞米松各20片。  
  • 第一天:化疗开始前,君凯(格拉斯琼)9毫克加入100毫升生理盐水中静脉滴注。紫杉醇210mg加入盐水500ml中(必须用玻璃瓶子,塑料瓶子可能会溶解),静脉点滴4小时(要求10点开始输注)
  • 应用3B输液器或输血器。化疗前30分钟,静脉入壶高舒达20mg或类似抑制胃酸药,苯海拉明40mg。  
  • 第二天:化疗开始前,君凯(格拉斯琼)9毫克加入100毫升生理盐水中静脉滴注。卡铂500mg加入5%葡萄糖溶液500ml中,静脉点滴4小时。  
  • 第三天,复查血常规、肝肾功。  
  • 休息3周,中间每周至少复查血常规2次,根据骨髓抑制程度注射升白药。
  • 紫杉醇配制时切忌用力摇晃,否则易出现过敏反应。请详细阅读说明书。
  • 紫杉醇应用可能有过敏反应出现,可为一型或四型变态反应。急性症状类似青霉素过敏,须抢救。
  • 紫杉醇应用可使血压升高,应用时应监测血压变化,必要时使用降压药。  
  • 其它紫杉醇的副反应如便秘,脱发等可针对病情予以处理。以上辅助用药可酌情换用其他类似药物。  
  • 如为首次应用紫杉醇,建议将30毫克紫杉醇加入250毫升生理盐水中缓慢滴注,确定没有过敏反应后将剩余紫杉醇按常规方法注射,以避免浪费药。


培美曲塞化疗方案
  • 化疗前常规检查血常规,肝肾功能,心电图。  
  • 化疗开始前七天,开始每日口服叶酸400ug,一直持续到最后一次化疗结束后21天。  
  • 化疗开始前七天内,任选一天给予维生素B12 1000ug肌肉注射一次。此后每三个周期肌肉注射一次维生素B12,可以与本药同一天进行。  
  • 化疗前一天,化疗当天,化疗后一天连续口服地塞米松每日两次,每次4mg。  
  • 化疗当天,培美曲塞(力比泰)500mg/m2稀释至100ml生理盐水,静脉注射10分钟以上。 顺铂75mg/m2加入5%葡萄糖500ml中,静脉点滴4小时。
  • 化疗前后可适量给予镇吐药物如昂丹司琼注射液等,如予以地塞米松5毫克与第一次镇吐药同时小壶滴注,止吐效果会明显增强。  
  • 输注顺铂时需按规定予以水化,碱化,利尿。  输注顺铂时,每天尿量需保持2000ml以上以保护肾功能。鼓励患者大量喝水以增加尿量。  
  • 本方案可每三周重复一次,但需定时复查血象及肝肾功能,可根据具体情况适量延长用药时间。


吉西他滨单药化疗方案
  • 化疗前常规检查血常规,肝肾功能,心电图。  
  • 第一天,第八天,第十五天,以下药物各静点一次:  
    • 格拉斯琼3毫克加入100毫升生理盐水中,化疗前静脉点滴。
    • 吉西他滨2.0g加入盐水100ml中,30分钟内快速静脉点滴。
    • 给予常规液体500毫升静点。  
    • 格拉斯琼3毫克加入100毫升生理盐水中,静脉点滴。
  • 化疗期间,每周至少复查血常规2次,根据骨髓抑制程度注射升白药。血常规需监测至化疗结束后2周。  
  • 吉西他滨副作用较少,主要有骨髓抑制发生,根据情况处理。特别注意其降低血小板作用较强,如血小板不低于4万,可观察,不需特殊处理。如低于4万且有出血倾向,可注射血小板刺激因子或输注血小板。


卡铂VP16(CE)化疗方案
  • 此化疗方案多针对小细胞癌。  
  • 第一天:卡铂500mg加入5%葡萄糖500ml中,静脉点滴4小时。化疗开始时,开始后3小时、6小时各加入枢丹8mg入壶。  
  • 第二天至第六天,每天足叶乙甙(VP16)100mg加入生理盐水500ml中,静脉点滴4小时。化疗开始时,开始后3小时、6小时各加入枢丹8mg入壶。  
  • 第七天,复查血常规、肝肾功。  
  • 休息2周,中间每周至少复查血常规2次,根据骨髓抑制程度注射升白药。
  • 第22--27天,同样方法静脉点滴卡铂及足叶乙甙。
  • 复查血常规及升白药用法同上。  
  • 以上药量可根据病人具体情况进行调整。  
  • 第一天可以将卡铂及足叶乙甙同一天使用,效果更好。
  • 副反应如便秘,脱发等可针对病情予以处理。  
  • 以上辅助用药可酌情换用其他类似药物,枢丹可以胃复安或类似药物代替。



“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:08:03 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-25 14:41 编辑

肺癌的分子靶向药物研究进展                    发表于:2011-10-14 22:23
http://www.haodf.com/zhuanjiaguandian/xuexingyang_533482403.htm


肺癌是发病率和死亡率最高的肿瘤,因为传统的化疗对非小细胞肺癌(NSCLC)效果有限,因此,迫切需要一些新的治疗方法。近年来分子靶向药物在肺癌方面取得一定成效,但由于价格昂贵,治疗方案也不成熟,有待深入研究。

分子靶向药物是在分子生物学、分子遗传学理论基础上出现的新药, 相对于传统化疗药物有很多优势, 形成了一门治疗肿瘤的新领域,主要包括细胞信号转导分子抑制剂、新生血管抑制剂、靶向端粒酶抑制剂以及针对肿瘤耐药的逆转剂等。

攻击肺癌细胞的靶点有多方面, 目前研究较成熟的主要有肿瘤细胞表面的靶点, 如单克隆抗体针对细胞膜分化相关抗原、细胞信号转导分子如表皮生长因子(EGF)及其受体(EGFR)和血管内皮生长因子(VEGF)及其受体上的酪氨酸激酶, 以及法尼基转移酶, 基质金属蛋白酶等[1]。

附表  肺癌新的分子靶向药物

药品名                            商品名                  作用靶点      
      
1.酪氨酸激酶抑制剂
吉非替尼片(Gefitinib)          易瑞沙(Iressa)            HER1/EGFR
厄罗替尼(Erlotinib)            他昔瓦(Tarceva)           HER1/EGFR  
拉帕替尼(Lapatinib)                                HER-2/EGFR双重抑制剂

2.新生血管抑制剂
贝伐单抗(Bevacizumab)        阿瓦斯丁(Avastin)          VEGF                       
西妥昔单抗(Cetuxmab)         爱必妥(Erbitux)            EGFR1        
泛尼特单抗(Panitumumab)      ABX-EGF                    EGFR1     
曲妥珠单抗(Trastuzumab)       赫赛汀(Herceptin)         EGFR2/Her-2/neu  
伊马替尼(Imatinib)            格列卫(Glivec)             CD117      
重组人血管内皮抑素(rh Endostatin)恩度(YH-16)               血管内皮素   
整合素拮抗剂(Vitaxin)                                      v整合素
基质金属蛋白酶抑制剂(Prinomastat)                       基质金属蛋白酶
内皮生长因子抑制剂(Thalidomide)    反应停                   内皮生长因子
法尼基转移酶抑制剂(Zarnestra)                               法尼基转移酶

3.维甲酸受体(RXR)抑制剂( Bexarotene)                      RXR

5.蛋白酶小体抑制剂(Bortezomib, Velcade )                      蛋白酶小体

6.环氧合酶2(Cox 2)抑制剂: 西乐葆(Celecoxib)              Cox 2

7.叶酸抑制剂: 培美曲塞二钠(Pemetrexed)阿莱慕塔&#174;(Alimta)  叶酸  

一、酪氨酸激酶抑制剂
NSCLC中EGFR和HER2基因通常被扩增, 受体基因过表达(EGFR表达率40 %~80%,其中鳞癌为 85%, 腺癌和大细胞癌为65%, 而小细胞肺癌罕见EGFR表达)。20%~30%NSCLC患者HER2过表达。这种过表达与预后不良、转移率高和总生存期短相关, 对判断术后疾病复发的高危性有意义。

酪氨酸激酶抑制剂的作用靶点是酪氨酸蛋白激酶(PTK),抑制细胞膜表面EGFR细胞内的众多酪氨酸激酶自磷酸化作用,阻断肿瘤细胞信号的传导,抑制肿瘤细胞的发展,诱导其凋亡。酪氨酸激酶抑制剂主要有EGFR酪氨酸激酶抑制剂和 Bcr-Abl(一种下调肿瘤激酶 )酪氨酸激酶抑制剂两种。

1.EGFR 酪氨酸激酶抑制剂:目前,针对信号传导分子-蛋白激酶而设计的EGFR酪氨酸激酶抑制剂主要有三种:吉非替尼、厄罗替尼和拉帕替尼。
(1)吉非替尼(Gefitinib),又称易瑞沙(Iressa)
吉非替尼是一种可以口服的苯胺喹唑啉类小分子化合物—EGFR酪氨酸激酶抑制剂(TKIs)。主要适用于治疗NSCLC和其他肿瘤如结直肠癌、头颈部癌、前列腺癌等。每日早餐后一小时口服250mg,可以达到治疗肿瘤的目的。增加剂量并不能提高疗效反而增加毒性。

据Ochs等在2004年ASCO会议报道,吉非替尼曾在全球开展慈善供药试验 (EAP),至2003年 7月有 21064例III/IV期化疗失败或无法耐受化疗的NSCLC患者服用吉非替尼,其中 9501例患者随访1年,20.1%的患者服药 6个月以上,全组患者中位生存期 5.3个月, 1 年生存率29.9%, 女性、亚洲人种、III期患者疗效较高。易瑞沙可单独用于铂类和紫杉醇类化疗失败的局部晚期或转移性NSCLC,不推荐使用联合化疗方案。最常见不良事件为腹泻、皮疹、痤疮、皮肤干燥、恶心和呕吐。

临床试验显示,包括中国在内的东方人群接受易瑞沙&#8482;治疗的结果非常显著。一项关于易瑞沙&#8482;延长肺癌患者生存期的国际多中心临床试验(ISEL)的初步分析结果显示,参加试验的东方人群的存活期中位数延长了4个月,几乎是其他受试组的两倍。可以使一些肺癌患者的肿瘤显著缩小,癌症症状明显缓解。该结果再次证实了以往的临床研究结果,确证易瑞沙&#8482;治疗东方病人的有效性。许多学者发现EGFR突变的NSCLC肿瘤细胞对TKIs相当敏感。在EGFR中体细胞突变最常发生于腺癌、非吸烟者,亚洲人和女性。检测EGFR突变成为判断TKIs是否敏感的重要指标。但作为EGFR信号传导介质的K-ras突变, 则与TKIs抵抗有关[2]。继发突变是 methionine取代激酶区790位上的threonine(T790M)。肿瘤细胞T790M的变异可导致对易瑞沙耐药,药物的溢出和蛋白结合也可阻止易瑞沙与靶位的结合。在ISEL临床试验中,未显示出易瑞沙比安慰剂组能延长患者生存期,因此现FDA已不再批准易瑞沙用于西方国家NSCLC的治疗 [3]。

(2)厄罗替尼(Erlotinib), 又称他昔瓦(TARCEVA&#8482; )
Erlotinib是一种1型EGFR /HER1酪氨酸激酶抑制剂,可口服,具有高效、高度特异的性质。Erlotinib为间二氮杂萘胺。适应于对至少一个化疗方案失败的局部晚期或转移的NSCLC患者的治疗。

剂量和用法:TARCEVA推荐的每日剂量为150 mg,在饭前1小时或饭后2小时服用。直到病情进展或出现无法接受的不良反应才停药。

从另外两项临床试验TALENT(n=1172)和TRIBUTE (n = 1059)得出的结论是[3]:将TARCEVA与含铂化疗方案[卡铂+泰素)或吉西他滨+顺铂联合用于局部晚期或转移的NSCLC 患者的一线治疗没有临床受益。因此,不推荐这类联合方案。TARCEVA有可能取代IRESSA而成为治疗NSCLC的三线治疗药物[3]。

通过一组随机、双盲、安慰剂对照的临床研究评价TARCEVA的安全性及疗效[4]。受试人数为731人,均为局部晚期或转移的NSCLC患者(对至少一个化疗方案失败),按2:1随机分配到接受每日一次口服TARCEVA 150 mg或安慰剂(488名为TARCEVA组,243名为安慰剂组),直到病情进展或出现无法接受的毒性才停药。结果显示,Tarceva中位生存期6.7月(安慰剂组4.7月),无进展生存期(PFS)9.9周,中位缓解时间34.3周,有效率(CR+PR) 8.9%(安慰剂<1%),1年生存率31.2%(安慰剂组21.5%),均较安慰剂组明显提高。TARCEVA对患者肿块EGFR阳性者(有效率为11.6%)和不吸烟者效果更好。

.不良反应:接受TARCEVA治疗的患者最常见的不良反应是皮疹、腹泻、食欲减退和疲乏各。皮疹发生的中位时间是8天,腹泻发生的中位时间是12天。出现皮疹患者的生存期比未出现皮疹患者的生存期明显延长(p<0.0001),表明皮疹可能是治疗有效的一个重要标志。

(3)拉帕替尼(Lapatinib):为EGFR和HER-2双重抑制剂
Lapatinib是苯胺喹唑啉类衍生物,是一种口服的ErbB-1 (EGFR) 和ErbB-2 (HER-2) 的双重抑制剂, 其对肿瘤增殖和生存的信号传导的抑制作用强于单一受体抑制剂。

在I期临床试验中发现,Lapatinib有很好的耐受性,最常见的不良反应是1~2级的消化道反应。临床试验发现,对gefitinib耐药的NSCLC也部分疗效。

在另外一组63名晚期或转移性NSCLC进行了II期 随机临床试验中[5],给予口服1500 mg/日(QD组)或500mg 2/日(BID组)。3级不良反应QD和BID组分别为腹泻(12%, 0%), 恶心(9%, 3%), 呕吐 (9%, 3%), 疲劳(3%, 3%), 呼吸困难(3%, 7%)和背痛(0%, 7%)。表明Lapatinib有很好的耐受性,副作用较轻。

剂量和用法:Lapatinib推荐的每日剂量为1250 mg,在饭前1小时或饭后2小时服用。

2.Ab1/Kit酪氨酸激酶抑制剂:
格列卫( Glivec),又名甲磺酸伊马替尼(Imatinib):选择性抑制少数相关的酪氨酸激酶,包括c-kit(CD117)、bcr-abl和PDGF受体。主要用于慢性粒细胞性白血病和胃肠间质瘤。

在一组II期临床试验中,29名c-kit阳性的SCLC随机分为两组,A组7例为以前治疗后进展<3个月的病人, B组22例为以前治疗后进展>3个月的病人。每组均给予Imatinib 400mg,2/日,28天一个周期。3级非血液性不良反应发生率为52%(主要为恶心,呕吐,呼吸困难,疲乏等)。两组MST分别为3.9和5.3个月,MTP分别为1和1.1个月。早期疾病进展率为29%, 早期死亡率为29%,患者拒绝率为42%。因此,Imatinib不适合c-kit表达阳性的 SCLC的治疗[6]。

二.新生血管抑制剂 
人体大部分肿瘤的生长和转移都依赖于病理条件下的血管生成, 因此抑制肿瘤介导的血管生成为肿瘤治疗提供了非细胞毒性的新途径。

1.贝伐单抗(Bevacizumab, rhu MAb-VEGF) ,商品名阿瓦斯汀(Avastin):是第一个重组的人源化抗血管内皮生长因子(VEGF)单抗, 能够结合并阻断 VEGF的作用, 从而发挥抗肿瘤活性。主要用于结直肠癌、乳腺癌、NSCLC和肾癌。

在一组随机、多中心II期临床试验中[7],99名新近诊断的IIIB期 (伴有胸腔积液)和IV期或复发性NSCLC 随机接受卡铂/紫杉醇治疗,或卡铂/紫杉醇+ bevacizumab治疗,均为每3周一次。结果显示,Bevacizumab (15 mg/kg) +卡铂/紫杉醇组与单纯化疗组比较明显增加反应率(RR31.4% vs 18.8%) 和平均疾病进展时间(MPT) (7.4 vs 4.2 月) 。存活时间也有所增加(14.2 vs 13.2月)。不良反应主要有高血压、血栓形成、蛋白尿和鼻衄。资料显示,鳞癌患者应用贝伐单抗有发生肺出血的危险,因此目前只用于非鳞癌的NSCLC。

在另外一组I/II期临床试验中[8], 40例复发性IIIb/IV期NSCLC病人(其中女性21例, 30例腺癌, 9例不吸烟,22例化疗2个以上疗程)同时接受贝伐单抗+ 厄罗替尼。结果部分有效率(PR)20.0%,稳定(SD)65.0%,II期试验中位总生存时间(MOS)为12.6个月,TTP为6.2个月。最常见的不良反应是轻至中度红斑、腹泻和蛋白尿。

2.西妥昔(Cetuximab), 商品名爱必妥Erbitux:是针对EGFR1的单抗,2004年2月由美国FDA批准用于发生转移的结肠癌,还可用于NSCLC。
Cetuximab是含人类IgG1 恒定区的人鼠嵌合性单克隆抗体,与EGFR结合可使细胞周期停滞,增加细胞周期由G1 到S期的重要调节因子p27的表达,促进肿瘤细胞凋亡,并通过下调VEGF等血管生成相关因子抑制肿瘤血管生成及转移。

Cetuximab与化疗合用具有协同作用。已证实Cetuximab与细胞毒性药物包括阿霉素、吉西他滨、顺铂、紫杉醇和拓扑替肯在广泛细胞系存在协同作用,并存在剂量依赖性,对NSCLC是安全、有效的。在以前的报道中[9],Cetuximab联合化疗的有效率在29%~53%。与多西紫杉醇联合治疗化疗耐药或复发的晚期EGFR阳性NSCLC患者,有效率为28%。

Rosell报道[10]在既往未接受化疗的ⅢB/Ⅳ期HER1/EGFR阳性NSCLC患者中,一组单独给予顺铂80mg/m2 d1+长春瑞宾25mg/m2 d1、8,3周重复;另一组在顺铂/长春瑞宾(剂量同前)化疗前使用Cetuximab 400mg/m2 第1周,随后每周250mg/m2;共62名患者参加,使用Cetuximab组有效率59%,而单独化疗组有效率仅32%。

在一项前瞻性研究中[11], 对铂类耐药的NSCLC患者每三周给予75 mg/m2 多西紫杉醇同时给予cetuximab初始剂量 400 mg/m2 ,继之维持计量 250 mg/m2。结果显示,RR为28%(包括1例CR),SD为 66%。疗效相当满意。

Cetuximab能增强放疗作用:Cetuximab和放疗亦有协同作用, 其作用机制可能为增加放疗敏感期 (G1,G2~S)肿瘤细胞、阻断放射诱导的DNA修复, 减少VEGF的产生, 减轻放射性损伤。美国东部肿瘤协作组(ECOG)的一项前瞻性研究提示, 与传统治疗相比, 对于不能手术切除的Ⅲ期NSCLC患者, Cetuximab联合超分割加速放疗将中位生存期由 13个月提高到20个月[12]。

Cetuximab联合其他生物靶向治疗,也可达到更好地治疗效果。蛋白印迹法检测发现, Cetuximab联合Erlotinib较单药明显降低活化的EGFR表达水平, 降低活化的有丝分裂活性蛋白激酶 (MAPK) , 增加对EGFR磷酸化的抑制,可使诱导凋亡的程度增加 3~4倍, 说明两药联合有协同作用 。

进一步研究发现, EGFR酪氨酸激酶抑制剂(TKIs)对EGFR单克隆抗体耐药的细胞系仍然有效, 其原因可能为EGFR下游信号传导不受Cetuximab影响, 而Gefitinib和Erlotinib仍能通过降低pMAPK和pakt的表达抑制EGFR下游信号, 结果表明, Cetuximab与Gefitinib、Erlotinib之间无交叉耐药[13], 故作用于EGFR不同分子结构域的抑制剂可以克服单一药物的局限性, 包括获得性耐药。

剂量及用法:推荐初始剂量为400mg/ m2(规格为100mg/50ml),滴注时间120分钟,滴速应控制在5ml/min以内。随后1周250mg,滴注时间不少于60分钟。使用前进行过敏试验。提前给予H1受体阻断剂。治疗终点为疾病进展或不能耐受的不良反应。

不良反应:主要Ⅲ或Ⅳ度毒副反应包括过敏、痤疮样皮疹、发热、乏力、恶心、肝酶升高等, 约 4%的患者出现Ⅲ或Ⅳ度过敏反应, 近11%的患者表现Ⅲ或Ⅳ度痤疮样皮疹。痤疮样皮疹是许多EGFR靶向治疗的特征性不良反应, 提示这部分患者的治疗效果可能好。

3.曲妥珠单抗(Trastuznmab),商品名赫赛汀(Herceptin):是针对EGFR2(或称erbB2,Her-2)等的人鼠嵌合抗体,1998年FDA批准用于卵巢癌和乳腺癌的治疗。

据报道[14],10~19%的NSCLC中HER2过度表达,腺癌高达30%,鳞癌达18%, 大细胞癌达11%。
一项II期临床试验显示,吉西他滨/顺铂联合Herceptin治疗NSCLC, 患者HER2 3+ 或基因复制阳性者,RR和MTP均明显延长,而HER2阴性者则无多大受益[15]。

4.Panitumumab(ABX-EGF)是一个完全人源化的 IgG2单克隆抗体, 作用于EGFR。单药治疗结直肠癌(CRC)、NSCLC、肾癌、前列腺癌、胰腺癌和食管癌有效,特别适合于晚期CRC[16]。目前尚缺乏NSCLC的大规模临床试验。

5.反应停(Thalidomide)
反应停通过抑制TNF  和转化因子 B而抑制肿瘤新生血管形成。体外试验证实, 反应停具有剂量依赖性地增加过氧化体增殖活化受体gamma (PPARγ) 蛋白和过氧化体增殖反应成分活性。因此, 反应停治疗大细胞癌(LCC)降低了细胞核因子 B的活性,增加凋亡和降低血管生成蛋白的表达[17]。

34例晚期NSCLC(3/4期6/28)II临床试验接受卡铂和Irinotecan治疗的同时加上反应停。23例可评估的病人中,PR22%, SD52%, MPT 3.6月, 19例病人死亡,MST 7.3月。最常见的不良反应是粒细胞减少、疲乏和恶心/呕吐[18]。

在另外一项I/II期临床试验中, 14例晚期复发性NSCLC接受了多西紫杉醇和反应停治疗。RR 14.2 % (1CR,1PR),SD 21.3%。TTP 11.5周,MST 3.4月。表明thalidomde(200 mg/day)结合docetaxel 75mg/m2 /3周是安全、有效地[19]。

6.法尼基转移酶抑制剂(Farnesyl Transferase Inhibitors , FTIs)
大约 30%的人类肿瘤与 RAS基因突变有关。RAS蛋白需要经过一系列的加工修饰才能定位于细胞膜内侧, 其中法尼基化是第一步也是其中最重要的一步。法尼基转移酶抑制剂干扰 RAS蛋白的法尼基化修饰, 可使 RAS基因激活的肿瘤生长受到抑制, 且对正常细胞无明显毒性。目前己经进入临床试验的有R115777(Zarnestra)、SCH-66336(Sarasar)、BMS-2 14662, L-778123等。

Zarnestra 和 L-778,123 作为FTIs能阻断 ras的信号传导,抑制细胞增殖和促进凋亡。在临床前试验中证实FTIs能抑制SCLC和NSCLC细胞生长。 在II期临床试验中,两药用于晚期初治的NSCLC和Zarnestra 用于复发的SCLC 均未达预期治疗效果,虽然Zarnestra治疗的少数病人中达SD。在后一组研究中发现>80%的病人法尼基转移酶有部分抑制,显示其一定的治疗效果。与其它靶向药物或化疗药结合应用的疗效正在进一步观察中[20]。

7.基质金属蛋白酶抑制剂(MMPIs): Prinomastat、BMS-275291
基质金属蛋白酶(MMPs)能降解细胞外蛋白,促进细胞生长、浸润、转移和血管形成。 在一组随机II期临床试验中,362名首次接受化疗的晚期NSCLC病人随机分为接受prinomastat 15 mg 或安慰剂,2/日。口服,联合吉西他滨+顺铂,结果显示两组RR、MTP和总生存率均为明显差别,证明prinomastat对NSCLC无明显作用[21]。

在另外的临床试验中, 也未观察到BMS-275291(广谱MMPIs)对NSCLC的疗效[22]。

病人在接受卡铂+紫杉醇治疗的同时,随机接受BMS-275291。774名患者BMS-275291组的OS,PFS和RR分别为8.6 月, 4.9月和25.8%,而安慰剂组分别为9.2 月, 5.3 月和33.7%。而BMS-275291组的不良反应较安慰剂组明显升高。

Marimastat 也是一种广谱MMPIs。在21例晚期NSCLC I期临床试验中,Marimastat 联合卡铂+紫杉醇治疗[23],结果PR57%,SD 19%。

在一组前瞻性、随机临床试验中,532例SCLC (266例接受 marimastat 和266安慰剂)。局限期占52%,广泛期占48%。 MPT在marimastat组为 4.3个月(安慰剂组为4.4个月),MST在 marimastat 和安慰剂组分别为9.3月 和9.7月。marimastat 3~6个月后生活质量呈恶化趋势。表明SCLC化疗后应用marimastat 并不能改善生存期,对生存质量亦有负面影响[24]。

8.重组人血管内皮抑制素恩度(YH-16)
血管内皮抑制素能抑制肿瘤血管增殖、血管形成和肿瘤生长。国内学者通过多中心期III期双盲、对照、前瞻性临床试验进一步证实了恩度的临床有效性[26]。 共有493例病理组织学证实的IIIB和 IV 期NSCLC。A组为NP方案 + YH-16 (n=326); YH-16 7.5mg·m-2(d1-14), NVB 25mg·m-2(d1,8)以及DDP 30mg·m-2(d2,3,4)。21d为1周期,共2~3周期。B组NP (n=167)。结果在完成治疗486的晚期患者中, 化疗结合恩度组总反应率(ORR)、临床效益率和TTP分别为35.4%、73.3%和6.3个月,均较安慰剂组明显提高(分别为19.5%、64.0%和3.6个月。  
                                          
两组的不良反应相似,均为3/4粒细胞减少症(均为28%左右)。

三.维甲酸受体(RXR)抑制剂
目前已知维生素 A衍生物—维甲酸 (Retinoic Acid, RA)具有多种生物学作用, 这些作用通过其受体介导。与 RA结合的受体分为两大类, 一类为胞质 RA结合蛋白(CRABP), 与 RA的代谢及转运有关; 另一类为核受体, 是一种转录调节因子, 它们又分为两类, 即维甲酸受体(Retinoic Acid Receptor, RAR)和维甲类 X受体 (Retinoic X Receptor, RXR), RAR和 RXR又分为 α、 β和 γ三种亚型。新发现的 RXR越来越引起人们的关注。RXR是核提取物中的一种辅因子,RXR s受体作为转录因子能调节许多细胞分化有关的基因。

Bexarotene:
Bexarotene与RXR结合,在动物模型中已经表现出抗细胞增殖活性,特别是鳞状细胞癌和支气管上皮来源的鳞状细胞癌。

在一组I/II期临床试验中[27],Bexarotene结合顺铂和长春瑞宾治疗NSCLC。43例IIIb合并胸水或IV期病人,接受bexarotene 结合顺铂和长春瑞宾治疗。bexarotene 剂量从150 mg/m2 到 600 mg/m2。在第一阶段bexarotene 400 mg/m2时,8/ 43有明显反应。 28例患者中25%进入第二阶段,有效率是25%,平均存活时间是14个月。 随访1,2,3年存活率分别为61% ,32%和30%。最常见的3 ~4级不良反应是高血脂症、白细胞减少、腹泻、恶心、肺炎和呼吸困难。

四.蛋白酶小体抑制剂:硼替佐米(Bortezomib),商品名万珂(Velcade)
Velcade(Bortezomib) 是一种小分子的水溶性二肽硼酸化合物,属于第 2代蛋白酶小体抑制剂,能抑制26S蛋白体酶,促进细胞凋亡[28]。主要用于复发性和难治性多发性骨髓瘤患者。也有一组II期临床试验万珂单药作为NSCLC二线治疗方案。研究显示,Bortezomib 通过抑制NF- B使肿瘤细胞对化疗药更加敏感。 但两药是序贯治疗还是同时治疗才能更好地发挥作用,有待深入研究。

在一组23例早期治疗(先前接受 1 个化疗方案)的NSCLC[29], 用bortezomib 单药治疗(剂量1.3~1.5 mg/m2 ,3周一次)。结果显示,1例患者PR,9例SD,反应率10/23。5个病人4个周期后仍有反应。

一组随机II 期临床试验中[30],比较了万珂1.3 mg/m2结合多西紫杉醇75 mg/m2 方案与单用万珂1.5mg/m2治疗晚期NSCLC。 两组的不良反应为恶心 (59% vs 35%), 疲劳(38% vs 48%), 腹泻 (38% vs 29%), 粒细胞减少症 (55% vs 3%),联合治疗组明显高于单用组。中期资料显示,单用组(n = 29)的PR为10.3% ,而联合组(n = 31)为16%。证明单用万珂对NSCLC有一定的疗效,而联合化疗效果更好。

五. v integrins抑制剂Vitaxin
Vitaxin是一种人源化的 v&#223;3 integrin单克隆抗体。在I期临床试验中,14例IV期肿瘤患者中,1例PR,7例SD。其中2例为NSCLC,1例无效,1例无法评价。表明该药在NSCLC中尚无法评定[31]。

六.以抗炎症为机制的抗肿瘤药
环氧合酶(Cyclooxygenase,Cox)是花生四烯酸转化为前列腺素的限速酶,Cox-2参与了肿瘤发展的诸多的基本环节如细胞凋亡、肿瘤浸润、血管再生和转移等,成为肿瘤大小、淋巴结转移和预后的重要指标。

研究表明,Cox-2在肺癌组织中高表达。选择性Cox2抑制剂可以抑制肿瘤的生长和转移,并可提高放疗和化疗的效果。

在一项I/II期临床试验中[32],49 名IIIB/IV患者一线给予顺铂/吉西他滨时结合应用Celecoxib。大多数患者收到了4个以上的周期的治疗。 绝大多数人有>50%的COX-2+肿瘤细胞。结果显示,Celecoxib 总生存率(OSR) 20.5%; 4个月的SD 43.2%。平均TTP 3+ months; 1年和2年生存率各为31.2% 和12.5%。

在另一项临床试验中[33],西乐堡联合paclitaxel/carboplatin治疗手术前的NSCLC,CR可达17%。

在另外一项II临床试验中[34],研究了gefitinib(250 mg,Qd)结合celecoxib(400 mg ,Bid) 治疗铂类耐药NSCLC。10例患者中PR 20%,SD 30%,证明两组联合对晚期NSCLC有效。

七.阿莱慕塔&#174;(Alimta&#174;)—注射用培美曲塞二钠(Pemetrexed,Eli Lilly and Company)
阿莱慕塔&#174;为含有吡咯嘧啶核的抗叶酸抗肿瘤药物,七水合培美曲塞二钠的化学名称为左旋谷氨酸,通过干扰对细胞复制至关重要的叶酸依赖的代谢途径而发挥作用。

在 NSCLC的II临床试验中[35],ALIMTA用于一线化疗进展3个月后或一线化疗3个月内进展者,81 名病人参加两组队列研究,A组先前含铂方案,B组无铂方案。用法: ALIMTA 500 mg/m2 10分钟内滴完,21天一个疗程。 结果在79名可评价的病人中,反应率为8.9%,A、B两组分别为4.5% 和14.1% 。平均生存时间分别为6.4个月和4.0个月。6个月的存活率为48%。TTP为2个月。主要的不良反应是骨髓抑制,35%的病人有3/4级粒细胞减少症。

在一组NSCLC的III临床试验中[36],采用单臂、对照、非双盲方法,研究571名复发性NSCLC随机分为ALIMTA&#174;治疗组和多西紫杉醇治疗组。平均生存时间分别为 8.3 个月和7.9个月,两组无明显差别。反应率分别为 9.1%和8.8%,两组亦无明显差异,表明ALIMTA&#174;单独治疗可达到与多烯紫杉醇相似的疗效。

两组单药、II期临床试验用于未治疗的NSCLC [37, 38]. 第一项研究中, 59名晚期病人接受pemetrexed ,500 mg/m2  21天一个疗程。ORR和平均生存时间(MST)分别为15.8% 和7.2个月。第二项研究中,33名早、中期病人接受pemetrexed ,600 mg/m2  21天一个疗程,ORR为23%,两组的毒性反应相似。 3/4级粒细胞减少症均为40%。

pemetrexed与顺铂、卡铂和吉西他滨结合治疗NSCLC取得更好的治疗效果。在两组II期临床试验中,67例病人接受pemetrexed+cisplatin [39,40]。两组的PR相似,均为45%,MST分别为9和11个月。50例接受pemetrexed +carboplatin [41],PR为28%,MST为14个月,3/4粒细胞减少症为34%。应用pemetrexed前1~2周及治疗过程中需补充VitB12 和叶酸。

60例晚期NSCLC(87%为IV期)给予吉西他滨结合pemetrexed,结果ORR 15.5%; SD 50.0%。总的平均生存时间(OS) 10.1 月, 1年和 2年生存率分别为42.6%和18.5%。PFS为5.0 月,平均反应持续时间3.3个月。表明两药结合有很好的耐受性,能明显延长1、2年生存率[42]。

参考文献
1.TJ Lynch , Adjei  AA., Bunn PA, et al. Novel Agents in the Treatment of Lung Cancer :Conference Summary Statement . Clin  Cancer Res   2004;10: 4199 S~4204 S
2.J&#228;nne PA, Engelman JA, Johnson BE. Epidermal Growth Factor Receptor Mutations in Non–Small-Cell Lung Cancer: Implications for Treatment and Tumor Biology.  J Clin Oncol, 2005;23(14):3227~3234
3.Siegel-Lakhai WS , Beijnen JH , Schellens JHM. Current Knowledge and Future Directions of the Selective Epidermal Growth Factor Receptor Inhibitors Erlotinib (Tarceva&#174;) and Gefitinib (Iressa&#174;). The Oncologist, 2005;10(8):579~589
4. Johnson JR, Cohen M, Sridhara R. Approval Summary for Erlotinib for Treatment of Patients with Locally Advanced or Metastatic Non–Small Cell Lung Cancer after Failure of at Least One Prior Chemotherapy Regimen . Clin Cancer Res  2005; 11:6414~6421
5.Ross HJ, Blumenschein GR, Dowlati A,et al. Preliminary safety results of a phase II trial comparing two schedules of lapatinib (GW572016) as first line therapy for advanced or metastatic non-small cell lung cancer . J Clin Oncol  2005: 23(16S): 7099
6.Dy GK, Miller AA, Mandrekar SJ,et al.A phase II trial of imatinib (ST1571) in patients with c-kit expressing relapsed small-cell lung cancer: a CALGB and NCCTG study. Ann Oncol  2005 16(11):1811~1816
7.Johnson DH,Fehrenbacher L, Novotny WF,et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone inpreviously untreated locally advanced or metastatic non-small cell lung cancer. J Clin Oncol  2004;22(11):2184~2191
8.Herbst RS, Johnson DH, Mininberg E,et al.Phase I/II Trial Evaluating the Anti-Vascular Endothelial Growth Factor Monoclonal Antibody Bevacizumab in Combination With the HER-1/Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Erlotinib for Patients With Recurrent Non–Small-Cell Lung Cancer. J  Clin Oncol , 2005;23(11):2544~2555
9.Govindan R. Cetuximab in Advanced Non-Small Cell Lung Cancer. Clin Cancer Res  2004; 10: 4241S~4244S
10.Rosell R, Daniel C, Ramlau R,et al.Randomized phase II study of cetuximab in combination with cisplatin (C) and vinorelbine (V) vs. CV alone in the first-line treatment of patients (pts) with epidermal growth factor receptor (EGFR)-expressing advanced non-small-cell lung cancer (NSCLC). J Clin Oncol  2004;22(14S): 7012
11.Kim ES, Mauer AM, Tran HT, et al. A phase II study of cetuximab, an epidermal growth factor receptor (EGFR) blocking antibody, in combination with docetaxel in chemotherapy refractory/resistant patients with advanced non-small cell lung cancer: Final report . Proc Am Soc Clin Oncol, 22: 642 2003
12.Belani CP,Wang W, Johnson DH, et al。Induction chemotherapy followed by standard thoracic radiotherapy vs hyperfractionated accelerated radiotherapy for patients with unresectable stageⅢA &B non small cell lung cancer:Phase Ⅲ studys of the Eastern Cooperative Oncology Group。Proc Am Soc Clin Oncol  2003;22:622
13、Huang S, Armstrong EA, Benavente S, et al. Dual agent molecular targeting of the epidermal frow factor receptor (EGFR):combining anti EGFR antibody with tyrosine kinase inhibitos. Cancer Res 2004;64:5355
14.Heinm&#246;ller  P, Gross C, Beyser K,et al. HER2 Status in Non-Small Cell Lung Cancer Results from Patient Screening for Enrollment to a Phase II Study of Herceptin。Clin Cancer Res . 2003; 9: 5238~5243
15.Gatzemeier U., Groth G., Hirsh V., Butts C., Van Zandwijk N., Shepherd F., Langer B., Rosso R. A randomised phase II study of gemcitabine/cisplatin alone and herceptin in patients with HER2-positive non-small cell lung cancer (NSCLC). ECCO, 2001;11: 173
16.Weiner LM, Belldegrun A, Rowinsky E,et al. Updated results from a dose and schedule study of Panitumumab (ABX-EGF) monotherapy, in patients with advanced solid malignancies. J Clin Oncol, 2005 ; 23(16S): 3059
17.DeCicco KL, Tanaka T, Andreola F,et al. The effect of thalidomide on non-small cell lung cancer (NSCLC) cell lines: possible involvement in the PPAR pathway. Carcinogenesis  2004 25(10):1805~1812
18.Miller AA, Case D, Atkins J,et al. Phase II study of carboplatin, irinotecan, and thalidomide in patients with advanced non-small cell lung cancer (NSCLC). J  Clin Oncol, 2004 ;22(14S): 7132
19.Seidler CW, Rooney J, Kodali D,et al. A phase I-II tri of docetaxel and daily thalidomide in patients with previously treated recurrent non-small cell lung cancer J  Clin  Oncol , 2004;22(14S): 7281
    20. Heymach JV, Johnson DH, Khuri FR, et al. Phase II study of the farnesyl transferase inhibitor R115777 in patients with sensitive relapse small cell lung cancer. Ann Oncol  2004; 15:1187~1193
21. Bissett D, O’Byrne KJ, von Pawel J, et al.Phase III Study of Matrix Metalloproteinase Inhibitor Prinomastat in Non–Small-Cell Lung Cancer. J Clin Oncol , 2005; 23(4):842~849
22.Rizvi NA, Humphrey JS,  Ness EA, et al. A Phase I Study of Oral BMS-275291, a Novel Nonhydroxamate Sheddase-Sparing Matrix Metalloproteinase Inhibitor, in Patients with Advanced or Metastatic Cancer . Clin Cancer Res    2004;10: 1963~1970
23.JR Goffin, Anderson IC, Supko JG, et al. Phase I Trial of the Matrix Metalloproteinase Inhibitor Marimastat Combined with Carboplatin and Paclitaxel in Patients with Advanced Non–Small Cell Lung Cancer.  Clin Cancer Res  2005; 11:3417~3424
24. Shepherd FA, Giaccone G, Seymour L, et al Prospective, randomized, double-blind, placebo-controlled trial of marimastat after response to first-line chemotherapy in patients with small-cell lung cancer: a trial of the National Cancer Institute of Canada-Clinical Trials Group and the European Organization for Research and Treatment of Cancer. J. Clin. Oncol., 2002;20: 4434~4439
25.Bissett D, O’Byrne K, von Pawel J, et al Phase III study of the matrix metalloproteinase inhibitor prinomastat in combination with gemcitabine and cisplatin in non-small cell lung cancer. Proc. Am. Soc. Clin. Oncol., 21: 296a 2002
26. Sun Y, Wang J, Liu Y, et al。 Results of phase III trial of rh-endostatin (YH-16) in advanced non-small cell lung cancer (NSCLC) patients 。J Clin Oncol  2005 ;23(16S): 7138
27.Khuri FR, Rigas JR, Figlin RA, et al. Multi-Institutional Phase I/II Trial of Oral Bexarotene in Combination With Cisplatin and Vinorelbine in Previously Untreated Patients With Advanced Non–Small-Cell Lung Cancer. J Clin Oncol  2001; 19(10): 2626~2637
28.Burger AM,Seth AK.The ubiquitin mediated protein degradation pathway in cancer: therapeutic implications.Eur J Cancer  2004;40:2217
29.Stevenson JP, Nho CW, Johnson SW et al. Effects of bortezomib (PS-341) on NF- B activation in peripheral blood mononuclear cells (PBMCs) of advanced non-small cell lung cancer (NSCLC) patients: a phase II/pharmacodynamic trial. Proc Am Soc Clin Oncol 2004;23:649a.
30.Fanucchi MP, Belt RJ, Fossella FV et al. Phase (ph) 2 study of bortezomib ± docetaxel in previously treated patients (pts) with advanced non-small cell lung cancer (NSCLC): preliminary results. Proc Am Soc Clin Oncol 2004;23:640a
31.Gutheil JC, Campbell TN, Pierce PR, et al. Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res 2000;6:3056~3061
32.Burton JD, El-Sayah D, Cherry M. Results of a phase I/II trial of carboplatin/gemcitabine plus celecoxib for first-line treatment of stage IIIB/IV non-small cell lung cancer (NSCLC). J  Clin Oncol, 2005;23(16S): 7250
33.Dubinett SM, Sharma, S, Huang, M, et al Cyclooxygenase-2 in lung cancer. Dannenberg, AJ DuBois, RN eds. COX-2: a new target for cancer prevention and treatment 2003,138-162 Karger. New York, NY
34.Gadgeel SM, Shehadeh NJ, Ruckdeschel JC. Gefitinib and celecoxib in patients with platinum refractory non-small cell lung cancer (NSCLC)。J  Clin Oncol , 2004;22(14S): 7094
35.Smit EF, Mattson K, von Pawel J,et al。ALIMTA&#174; (pemetrexed disodium) as second-line treatment of non-small-cell lung cancer: a phase II study。 Ann Oncol 2003;14:455-460
36.Hanna N, Shepherd FA, Fossella FV et al. Randomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapy. J Clin Oncol 2004;22:1589–1597
37.Clarke SJ, Abratt R, Goedhals L et al. Phase II trial of pemetrexed disodium (ALIMTA, LY231514) in chemotherapy-naive patients with advanced non-small-cell lung cancer. Ann Oncol 2002;13:737~741
38.Rusthoven JJ, Eisenhauer E, Butts C et al. Multitargeted antifolate LY231514 as first-line chemotherapy for patients with advanced non-small-cell lung cancer: a phase II study. National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 1999;17:1194~1199
39.Shepherd FA, Dancey J, Arnold A et al. Phase II study of pemetrexed disodium, a multitargeted antifolate, and cisplatin as first-line therapy in patients with advanced nonsmall cell lung carcinoma: a study of the National Cancer Institute of Canada Clinical Trials Group. Cancer 2001;92:595~600
40.Manegold C, Gatzemeier U, von Pawel J et al. Front-line treatment of advanced non-small-cell lung cancer with MTA (LY231514, pemetrexed disodium, ALIMTA) and cisplatin: a multicenter phase II trial. Ann Oncol 2000;11:435~440
41.Koshy S, Herbst RS, Obasaju CK et al. A phase II trial of pemetrexed (P) plus carboplatin (C) in patients (pts) with advanced non-small-cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 2004;23:631a
42.Monnerat C, Le Chevalier T, Kelly K et al. Phase II study of pemetrexed-gemcitabine combination in patients with advanced-stage non-small cell lung cancer. Clin Cancer Res 2004;10:5439~5446
   
“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:10:28 | 显示全部楼层 来自: 美国

阿瓦斯丁和恩度

本帖最后由 荷花池荒岛 于 2014-8-31 08:46 编辑



1)http://www.ay91.com/Cancer/Doctor/lllf/84360.htm
     。。。。。。
     2、肺癌常用的靶向治疗有几种?代表药物是什么?
    根据药物的性质和作用靶点,在肺癌治疗中常用的靶向治疗主要有以下两类。

    第一类是针对肿瘤血管的,包括抗血管内皮细胞生长因子的单克隆抗体和血管内皮抑素。前者的代表药物是阿瓦斯丁(Avastin,贝伐单抗),后者的代表药物是国产的恩度(Endostar)。

    第二是作用于肿瘤细胞信号传导通路的小分子物质,比较常见的是肿瘤表皮生长因子受体抑制剂,代表药物包括易瑞沙(吉非替尼)、特罗凯(厄罗替尼)等,临床应用较为普遍。
    。。。。。。
    4、靶向治疗常见的不良反应和注意事项
    易瑞莎和特罗凯是表皮生长因子受体(EGFR)酪氨酸激酶抑制剂,常见的毒副反应是腹泻、痤疮样皮疹、瘙痒、皮肤干燥,发生率20%以上,恶心呕吐发生率15%,一般见于服药后一个月内,通常是可逆性的。有极少一部分病人在接受以上两种药物后出现间质性肺病,,一旦确诊应当立即停药。贝伐单抗常见不良反应时高血压、蛋白尿、血栓证等,一般不影响治疗,最为严重的不良反应是肿瘤相关性出血,如咯血或呕血,因此禁用于有严重出血倾向及肺鳞癌患者。
    。。。。。。


2)http://www.haodf.com/zhuanjiaguandian/xuexingyang_533482403.htm
    。。。。。。
二.新生血管抑制剂 
人体大部分肿瘤的生长和转移都依赖于病理条件下的血管生成, 因此抑制肿瘤介导的血管生成为肿瘤治疗提供了非细胞毒性的新途径。
1.贝伐单抗(Bevacizumab, rhu MAb-VEGF) ,商品名阿瓦斯汀(Avastin):是第一个重组的人源化抗血管内皮生长因子(VEGF)单抗, 能够结合并阻断 VEGF的作用, 从而发挥抗肿瘤活性。主要用于结直肠癌、乳腺癌、NSCLC和肾癌。

在一组随机、多中心II期临床试验中[7],99名新近诊断的IIIB期 (伴有胸腔积液)和IV期或复发性NSCLC 随机接受卡铂/紫杉醇治疗,或卡铂/紫杉醇+ bevacizumab治疗,均为每3周一次。结果显示,Bevacizumab (15 mg/kg) +卡铂/紫杉醇组与单纯化疗组比较明显增加反应率(RR31.4% vs 18.8%) 和平均疾病进展时间(MPT) (7.4 vs 4.2 月) 。存活时间也有所增加(14.2 vs 13.2月)。不良反应主要有高血压、血栓形成、蛋白尿和鼻衄。资料显示,鳞癌患者应用贝伐单抗有发生肺出血的危险,因此目前只用于非鳞癌的NSCLC。

在另外一组I/II期临床试验中[8], 40例复发性IIIb/IV期NSCLC病人(其中女性21例, 30例腺癌, 9例不吸烟,22例化疗2个以上疗程)同时接受贝伐单抗+ 厄罗替尼。结果部分有效率(PR)20.0%,稳定(SD)65.0%,II期试验中位总生存时间(MOS)为12.6个月,TTP为6.2个月。最常见的不良反应是轻至中度红斑、腹泻和蛋白尿。
    。。。。。。
8.重组人血管内皮抑制素恩度(YH-16)
血管内皮抑制素能抑制肿瘤血管增殖、血管形成和肿瘤生长。国内学者通过多中心期III期双盲、对照、前瞻性临床试验进一步证实了恩度的临床有效性[26]。 共有493例病理组织学证实的IIIB和 IV 期NSCLC。A组为NP方案 + YH-16 (n=326); YH-16 7.5mg·m-2(d1-14), NVB 25mg·m-2(d1,8)以及DDP 30mg·m-2(d2,3,4)。21d为1周期,共2~3周期。B组NP (n=167)。结果在完成治疗486的晚期患者中, 化疗结合恩度组总反应率(ORR)、临床效益率和TTP分别为35.4%、73.3%和6.3个月,均较安慰剂组明显提高(分别为19.5%、64.0%和3.6个月。                                             
两组的不良反应相似,均为3/4粒细胞减少症(均为28%左右)。
   。。。。。。


3)恩度  http://51qiji.com/thread-17081-3-1.html


4)http://www.yuaigongwu.com/thread-15361-1-2.html   3楼、6楼
恩度无效,贝伐有效临床上存在的。恩度作用靶点广泛,作用靶点广泛,可以下调促血管生成和促进肿瘤增殖转移因子的表达,如低氧诱导因子(HIF)、血管内皮生长因子(VEGF)、血小板源性生长因子(PDGF)、成纤维细胞生长因子(FGF)、金属蛋白酶(MMP)、整合素(integrin)等,最终通过抑制血管内皮的生长,但是效力不如贝伐单抗强。贝伐单抗是结合VEGF-A亚型结合,以此阻断其与受体VEGFR-2(Flt-1/KDR)结合。

"恩度是血管内皮抑素,一种天然存在的抑制血管生成的物质;贝伐单抗是anti-VEGF单抗,清除血管内皮生长因子。虽然都是抑制血管生成,但分子机制不同,贝伐单抗效果更好,欧美人主要用贝伐单抗,恩度这个药只在中国获批了。贝伐单抗 紫杉醇 卡铂是治疗非鳞状非小细胞肺癌的一种标准方案,但不要对贝伐单抗抱有太大的期望,化疗中增加贝伐单抗与单纯化疗的中位生存期是12.3个月 vs 10.3个月。"


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:10:57 | 显示全部楼层 来自: 美国

学习治疗帖

本帖最后由 荷花池荒岛 于 2016-9-6 12:31 编辑

1)今生的等待  http://app.yuaigongwu.com/forum. ... &extra=page%3D1

2)echo_mayi http://www.yuaigongwu.com/forum. ... amp;_dsign=dd402f48

3)jiangfei  http://wwsw.yuaigongwu.com/forum ... amp;_dsign=578d7e26

4)bishop_cn http://qqq.yuaigongwu.com/forum. ... amp;_dsign=07a6e2fe

5)好梦成真 http://www.yuaigongwu.com/forum. ... &extra=page%3D4

6)wendy.0321  http://download.yuaigongwu.com/f ... amp;_dsign=2c5921a0

7)绿茶 http://app.yuaigongwu.com/forum.php?mod=viewthread&tid=396

8)老公天天开心  http://www.yuaigongwu.com/forum. ... amp;_dsign=80f3c07a

9)老马  http://www.yuaigongwu.com/forum. ... amp;_dsign=d611efa2

10)never_land  http://www.yuaigongwu.com/forum. ... amp;_dsign=d4ba4372

11) 绿妖之妖  http://www.yuaigongwu.com/forum. ... amp;_dsign=faaa4eae

12)镇江小杨  http://w3w.yuaigongwu.com/forum. ... amp;_dsign=ae85af56

13)与爱有别  http://www.yuaigongwu.com/forum. ... amp;_dsign=ae0cda21

14)ak780401  http://www.yuaigongwu.com/forum. ... age%3D14&page=1

15)潇湘风雨  http://www.yuaigongwu.com/thread-13915-1-3.html

16)wlxkxgq  http://wwsw.yuaigongwu.com/forum.php?mod=viewthread&tid=13012

17)爸爸最棒  http://w3w.yuaigongwu.com/forum. ... amp;_dsign=39f3d7fc

18)简单的微笑  http://wwsw.yuaigongwu.com/forum.php?mod=viewthread&tid=12212

19)Alice  http://forum.yuaigongwu.com/thread-5572-1-1.html?_dsign=e3b94b94

20)lb9702 http://www.yuaigongwu.com/forum. ... &extra=page%3D1


探锁理论实践帖:
n95e71的一位靶向受益者,对[探锁的心]理论的看法:
http://w3w.yuaigongwu.com/forum.php?mod=viewthread&tid=22876

治疗肺腺癌---浙江4期肺腺癌治疗征程
http://www.yuaigongwu.com/forum.php?mod=viewthread&tid=24732

梦想飞翔的探锁的心 危险信号理论 肺鳞癌 治疗实践:
http://w3w.yuaigongwu.com/forum. ... amp;_dsign=6c7ecf6f

ufo8808的熊猫家关于实施探锁理论的病例治疗贴:
http://w3w.yuaigongwu.com/forum. ... &extra=page%3D2

胶质瘤家属2014:
http://bbs.tianya.cn/post-100-1904211-1.shtml

猫猫家肺腺癌治疗记录
http://www.yuaigongwu.com/thread-28570-1-1.html?_dsign=915e4b7b

JINANGLL  http://qwww.yuaigongwu.com/forum ... amp;_dsign=5293e9a3






“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:11:12 | 显示全部楼层 来自: 美国

服用易瑞沙、特罗凯不能吃葡萄柚

本帖最后由 荷花池荒岛 于 2014-9-8 03:46 编辑

     
     服用特罗凯(厄洛替尼)期间,并非忌吃柚子和葡萄,而是特指一种叫做“葡萄柚”的水果或其果汁,会引起很多经过肝脏代谢的口服药物血液浓度提高。因此,建议葡萄柚(或葡萄柚汁)尽可能不要与药物同时服用,而服用葡萄柚(或葡萄柚汁)期间(三天之内),应该减少口服药物用量,以免发生不良反应。

     葡萄柚(grapefruit)亦名西柚或胡柚,主要被肝脏细胞色素P450酶系统中CYP3A4酶代谢,大量研究表明它同时也能抑制CYP3A4酶的活性,从而抑制药物的氧化代谢。因此,许多被CYP3A4酶代谢的口服药物,都会因CYP3A4酶的活性被抑制而使血液浓度提高,如口服的厄洛替尼片(特罗凯)、吉非替尼(易瑞沙)、伊马替尼(格列卫)、多潘立酮(吗丁啉)、西沙必利(普瑞博思)、辛伐他汀(舒降之)、洛伐他汀(美降之)、非洛地平(波依定)、尼卡地平、华法令、氨茶碱、他克莫司(普乐可复)、环孢菌素(新山地明)、瑞格列奈(诺和龙)、卡马西平、三唑仑、咪达唑仑(多美康)等。

     此外,能够抑制CYP3A4酶活性的除了葡萄柚(或葡萄柚汁)之外,还包括环丙沙星、克拉霉素、泰利霉素、三乙酰竹桃霉素、伊曲康唑、酮康唑、奈法唑酮、伏立康唑、阿扎那韦、茚地那韦、奈非那韦、利托那韦、沙奎那韦等,具体请向医生咨询。


http://blog.sina.com.cn/s/blog_69ecff150102ux00.html


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:17:15 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-9-8 04:12 编辑

http://app.yuaigongwu.com/forum. ... &extra=page%3D1

NSCLC恶性胸水治疗01.PNG
NSCLC恶性胸水治疗02.PNG
NSCLC恶性胸水治疗03.PNG
“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:17:25 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-9-8 04:39 编辑

1)
疾病进展(PD, Progressive Disease) 靶病灶最大径之和至少增加≥20%,或出现新病灶    

部分缓解(PR, Partial Response) 靶病灶最大径之和减少≥30%,至少维持4周  

疾病稳定(SD, Stable Disease) 靶病灶最大径之和缩小未达PR,或增大未达PD  

完全缓解(CR, Complete Response) 所有靶病灶消失,无新病灶出现,且肿瘤标志物正常,至少维持4周

客观缓解率(ORR, Objective Response Rate) 指肿瘤缩小达到一定量并且保持一定时间的病人的比例,包括CR+PR的病例

疾病控制率(DCR,Disease Control Rate):CR+PR+SD

无进展生存期(PFS,Progress Free Survival) 是指从随机化开始至出现疾病进展或死亡的时间

疾病进展时间(TTP,Time To Progress) 指从随机分组开始到肿瘤客观进展的时间

总生存期(OS, Overall Survival) 指从随机化开始至因任何原因引起死亡的时间

标靶治疗(TT, Targeted Therapy)

http://www.haodf.com/zhuanjiaguandian/seastone2008_836932735.htm


2)
Adenosine TriPhosphate (ATP)                          三磷酸腺苷

Epidermal Growth Factor Receptor (EGFR)         表皮生长因子受体

Hepatocyte Growth Factor(HGF)                       肝细胞生长因子

Human Epidermal growth actor Receptor(HER)  人体表皮生长因子受体

Interstitial Lung Disease (ILD)                         间质性肺疾病

mammalian Target Of Rrapamycin(mTOR)   哺乳动物的雷帕霉素靶

Maximum-Tolerated Dose (MTD)                      最大耐受剂量

Minimum Inhibitory Concentration (MIC)           最低抑菌浓度

Phosphatidylinositol 3-Kinase(PI3K)                   磷脂酰肌醇3 - 激酶

Tyrosine Kinase (TK)                                      酪氨酸激酶

Tyrosine Kinase Inhibitor (TKI)                        酪氨酸激酶抑制剂

Vascular Endothelial Growth Factor Receptor(VEGFR) 血管内皮生长因子受体




3)保健品:
CoQ10                                                                                心脏
Digestive Enzyme Complex                          消化酶                  胃
Gamma Vitamin E  
Ginkgo Biloba                                           银杏胶囊                老年痴呆
Glutathione                                              谷胱甘肽                免疫力
Lactoferrin                                               乳铁蛋白                开胃、免疫力
Lipoic Acid                                               硫辛酸                   免疫力
Milk Thistle                                               水飞蓟宾                肝
Turmeric Curcumin                                    姜黄素(注:要买含黑胡椒black pepper的。)


4)心力衰竭治疗药物:
比索洛尔(默克的康忻)
卡维地洛(罗氏的达利全)
美托洛尔(阿斯利康的倍他乐克)


5)降心率药物:
伊伐布雷定(Procoralan)


6)心力衰竭辅助药物:
辅酶Q10(能气朗)
钾的混合液(GIK溶液)
门冬氨酸钾镁片(潘南金)
葡萄糖
盐酸曲美他嗪片(万爽力)
胰岛素
左卡尼汀(左旋肉毒碱 L-carnitine)

注:万爽力和左卡尼汀具有拮抗作用,不能联合。左卡尼汀与抗高血压药赖诺普利(Lisinopril)也不能合用。


7)其它药物:
间质性肺炎(富露施 )
脑梗、老年痴呆(银杏胶囊)
血糖(二甲双胍)


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-25 02:17:49 | 显示全部楼层 来自: 美国

替莫唑胺

本帖最后由 荷花池荒岛 于 2014-9-8 12:38 编辑

1)替莫唑胺中文简要说明书
http://www.yiruisha.com/SMS/%E6% ... %89%88%EF%BC%89.pdf


2)http://oncol.dxy.cn/article/9829?trace=related
。。。。。。
替莫唑胺同步放化疗治疗脑转移疗效显好

替莫唑胺(TMZ)是一种新型的咪唑四嗪类烷化剂,口服吸收完全,生物利用度高,可透过血脑屏障,其在脑胶质瘤中的疗效已获证实,在肺癌等实体瘤脑转移中的疗效亦初显端倪。

一项意大利的Ⅱ期研究评价了 TMZ 单药标准治疗(150~200mg/mm/d,d1~5, 每 28 天重复)作为非小细胞肺癌(NSCLC)脑转移挽救治疗的疗效。研究入组 30 例 NSCLC 患者,结果显示,脑转移灶客观缓解率(ORR)为 10%,总体的至疾病进展时间(TTP)和总生存期(OS)分别为 3.6 个月和 6 个月,其中获得客观缓解的患者 TTP 和 OS 分别达到了 11~19 个月和 14~24 个月。

另外一项Ⅱ期研究采用 TMZ 每日低剂量(75mg/mm/d,d1~21,每 28 天重复)的方式治疗复发难治的 NSCLC,其中合并脑转移的患者占 39%,疾病控制率(DCR)为 16.2%,TTP 和 OS 分别为 2.4 个月和 3.3 个月。这两项临床研究均显示 TMZ 作为 NSCLC 脑转移二线以上的治疗有一定疗效,值得开展Ⅲ期临床研究。

TMZ 联合放疗治疗脑转移同样显示出较好的疗效。法国一项Ⅱ期临床研究对 50 例脑转移的 NSCLC 患者采用 TMZ 联合顺铂化疗序贯全脑放疗,结果显示 ORR 为 16%,TTP 和 OS 分别为 2.3 个月和 5 个月。而另外两项关于 TMZ 同期放化疗的Ⅱ期临床研究显示,ORR 达到了 45%~57.6%,OS 更是达到了 12~13 个月。这几项研究提示同步放化疗可能优于序贯化放疗或单一化疗。

另外,TMZ 对于脑转移的预防可能也有一定的作用。有研究显示,TMZ 联合拓扑替康治疗 NSCLC,仅有 8%(3/37 例)的患者最终出现了脑转移,远远低于其他文献报道的 50% 的脑转移发生率,提示 TMZ 可能具有潜在的预防脑转移的作用。

TMZ 在肺癌脑转移治疗所显示的作用,值得开展进一步的临床试验。基于 TMZ 在脑瘤和各种脑转移中的表现,2009 版 NCCN 指南将其作为脑部肿瘤的化疗选择之一予以推荐。
。。。。。。


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”

发表回复

您需要登录后才可以回帖 登录 | 立即注册

本版积分规则

  • 回复
  • 转播
  • 评分
  • 分享
帮助中心
网友中心
购买须知
支付方式
服务支持
资源下载
售后服务
定制流程
关于我们
关于我们
友情链接
联系我们
关注我们
官方微博
官方空间
微信公号
快速回复 返回顶部 返回列表