• 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

T790M突变和cMet扩增

    [复制链接]
192402 187 老马 发表于 2012-12-27 12:54:50 |

马上注册,结交更多好友,享用更多功能,让你轻松玩转社区。

您需要 登录 才可以下载或查看,没有账号?立即注册

x
本帖最后由 老马 于 2013-2-25 08:04 编辑

1.        T790M突变
1.1 发生机理
L858R氨基酸改变(21突变)或L747A-A750缺失(19突变)这两种突变形式使EGFR总是处于被激活的状态。易瑞沙/特罗凯可以抑制EGFR酪氨酸激酶磷酸化及其底物磷酸化,从而使EGFR处于非激活的状态,因而抑制肿瘤生长。
一些研究结果显示在易瑞沙/特罗凯耐药的患者中,相当一部分能检测到T790M突变。这种突变能使表皮生长因子受体(EGFR)与ATP的亲和力增加,因而可逆性TKI难以继续抑制EGFR的磷酸化。
对吉非替尼敏感的PC-9细胞经由不同浓度的吉非替尼处理后,逐渐显示出对吉非替尼耐药,应用测序及突变富集PCR法可以在第44天时检测到T790M突变。提示吉非替尼暴露能逐渐诱导出T790M突变。而在280例患者样本中应用突变富集PCR法能更敏感地检测到T790M突变,而且部分吉非替尼治疗前的患者存在T790M突变的微小克隆,经吉非替尼治疗后,敏感克隆被杀死,而含有T790M突变的耐药克隆得以保留下来。非小细胞肺癌经吉非替尼治疗后出现T790M突变,可能存在两种机制,一种是由于吉非替尼慢性暴露诱导EGFR20号外显子2369核苷酸发生C→T错义突变;另一种是肿瘤中原本存在着T790M突变的微小克隆,经吉非替尼选择后,耐药克隆得到扩增。
对外显子20的扩增产物进行亚克隆分析发现,这种新的突变是一个碱基对发生从胞嘧啶核苷(C)到胸腺嘧啶核苷(T)的改变,T790M这种突变使苏氨酸(Threonie)变成蛋氨酸(Methionie),蛋氨酸大的侧链构型阻碍了吉非替尼的结合,同时,使EGFR与ATP的亲和力明显增加,使EGFR重新处于被激活状态。
研究显示约3.6%未经治疗的NSCLC患者肿瘤组织标本中T790M突变阳性,而对吉非替尼或厄洛替尼耐药的NSCLC患者其阳性表达率为43%-50%。说明T790M突变机率低,但却与TKI耐药显著相关。
不可逆EGFR酪氨酸激酶抑制剂,如BIBW2992、和PF00299804等对T790M突变的细胞仍有效。
1.JPG
1.2 临床特征
T790M 突变多见于肺组织/胸膜和淋巴结,进展缓慢,较少发生于远处转移。T790M 突变的易瑞沙/特罗凯耐药病人的平均进展生存期(PPS)没有发生T790M 突变的病人更长(19个月对12个月),总生存期(OS)也更长(39个月对26个月)。
2.JPG
3.JPG
4.JPG
5.JPG
2. cMet扩增
2.1 发生机理
MET基因位于染色体7q31,编码分子为190KD的跨膜糖蛋白,属酪氨酸激酶生长因子受体家族成员,其蛋白产物为肝细胞生长因子受体(HGF),与细胞的增殖能力有关。研究显示MET基因扩增能激活ErbB3/PI3K/AKT信号途径,引发对EGFR激酶抑制剂的耐药性。2007年Engelman 等在其建立的吉非替尼耐药的细胞系中检测到MET基因的扩增,并通过对MET信号通路的阻断恢复对吉非替尼的敏感性。在其检测的18例对吉非替尼或厄洛替尼耐药的NSCLC标本中,有4例(22%)检测到了MET基因的扩增;有8例患者在服用靶向药 物治疗前后均检测了MET扩增情况,其中2例疗前无MET基因扩增的患者在接受靶向治疗后出现扩增;有1例对TKI继发耐药患者同时检测到MET基因扩增及T790M突变,另1例患者在不同转移灶中分别检测到T790M突变及MET基因扩增。另有报道,20%NSCLC TKI耐药与c-MET基因扩增有关,其发生与T790M的存在无相关性,通过抑制MET基因的扩增,可以使肺癌靶向治疗的有效率从71%提高到93%。
吴一龙等获得55例术后非小细胞肺癌(NSCLC)的肿瘤组织(基线组)以及23例对TKIs耐药的肿瘤组织(耐药组)后,通过激光显微切割筛选癌细胞后提取基因组DNA,实时荧光定量PCR TaqMan探针法检测所有标本的c-MET基因的拷贝数。结果:1.基线组和耐药组的临床病理特征均与c-MET基因的扩增无关。2.基线组中c-MET基因扩增阳性率为5.5% (3/55);耐药组的c-MET基因扩增阳性率为21.7% (5/23)。
6.JPG
耐药图.JPG
5684703_1361313967k.jpg

点评

没有老马,前沿信息都变得不前沿了!  发表于 2017-10-5 14:31
看完只能感慨:高手在民间!  发表于 2017-2-13 17:05

本帖被以下淘专辑推荐:

个人公众号:treeofhope

210条精彩回复,最后回复于 2022-7-26 21:37

老马  博士一年级 发表于 2012-12-27 12:55:39 | 显示全部楼层 来自: 浙江温州
3.易瑞沙/特罗凯耐药对策
7.JPG
个人公众号:treeofhope
老马  博士一年级 发表于 2012-12-27 12:56:08 | 显示全部楼层 来自: 浙江温州
本帖最后由 老马 于 2013-2-14 11:41 编辑

Combined EGFR/MET or EGFR/HSP90 inhibition is effective in the treatment of lung cancers co-driven by mutant EGFR containing T790M and MET
http://cancerres.aacrjournals.or ... AN-11-3720.abstract
Tyrosine kinase inhibitors (TKIs) that target the epidermal growth factor receptor (EGFR) are effective in most NSCLC patients whose tumors harbor activating EGFR kinase domain mutations. Unfortunately, acquired resistance eventually emerges in these chronically treated cancers. Two of the most common mechanisms of acquired resistance to TKIs seen clinically are the acquisition of a secondary "gatekeeper" T790M EGFR mutation that increases the affinity of mutant EGFR for ATP and activation of MET to offset the loss of EGFR signaling. Although up to one-third of patient tumors resistant to reversible EGFR TKIs harbor concurrent T790M mutation and MET amplification, potential therapies for these tumors have not been modeled in vivo. In this study, we developed a preclinical platform to evaluate potential therapies by generating transgenic mouse lung cancer models expressing EGFR-mutant Del19-T790M or L858R-T790M, each with concurrent MET overexpression. We found that monotherapy targeting EGFR or MET alone did not produce significant tumor regression. In contrast, combination therapies targeting EGFR and MET simultaneously were highly efficacious against EGFR TKI resistant tumors co-driven by Del19-T790M or L858R-T790M and MET. Our findings therefore provide an in vivo model of intrinsic resistance to reversible TKIs and offer preclinical proof of principle that combination targeting of EGFR and MET may benefit patients with NSCLC.

个人公众号:treeofhope
老马  博士一年级 发表于 2012-12-27 12:56:12 | 显示全部楼层 来自: 浙江温州
本帖最后由 老马 于 2013-2-18 00:46 编辑

Don’t Kick the TKI (When EGFR+ Lung Cancer Progresses)
http://www.medscape.com/viewarticle/757942
Hello. This is Mark Kris from Memorial Sloan-Kettering Cancer Center in New York. I would like to talk more about disease progression in lung cancer. This is a common issue. We deal with it every day, but very little critical evaluation about the definition and assignment of disease progression has been done. Clinical scenarios differ. In the last commentary, I spoke about the use of progression criteria in patients receiving a traditional or standard cytotoxic regimen.

What about patients receiving a tyrosine kinase inhibitor (TKI)? First, we have to remember that these patients have a different illness. Their form of lung cancer is somewhat different from others. Their cancer cells are addicted to the signaling of EGFR (epidermal growth factor receptor) or of ALK (anaplastic lymphoma kinase), the 2 most common situations we have in lung cancer.

These tumors are made up of cells of various molecular wiring. Some of the cells have a wild-type gene or at least 1 wild-type allele. Others have mutated genes. Even at the time of resistance, only a small number of genes have the second site or resistance mutation. The truth is that when you can first identify those resistance mutations, they are a small number of all of the alleles of EGFR. If you stop the EGFR-TKI, the vast number of cells are still sensitive, and they will likely grow even faster.

It turns out from laboratory experiments and observations in patients that the second site mutations with EGFR (the T79DM mutation) impart a somewhat slower growth than the sensitivity mutation-driven tumors. With time, if you were to stop an EGFR-TKI, the resistant cells would continue to grow slowly. They represent a minority of the cells in that tumor. The cells that are still sensitive to the TKI grow very rapidly. Many of you have seen the so-called "flare phenomenon," which is an example of that rapid growth.

Knowing this biology, which is clearly something different from what we were all taught in oncology school, what do you do? Just as in cytotoxic chemotherapy, when a tumor lesion becomes symptomatic, when clearly a new lesion appears, and particularly a new disease site, that is probably a time to modify therapy. The modification in this situation is adding something to the EGFR-TKI.

What about situations when you have only growth in the indicator lesion? Please remember that if you are using RECIST criteria, it is a 20% change from the smallest size of the tumor. If an 8-cm tumor decreased to 2 cm and then suddenly grew to 2.4 cm (just about within the limits of detection and reproducibility of a CT scanner), that is RECIST progression.

We know from these patients that they have a different biology and sometimes a very different clinical course. Many of you who treat patients in this situation have seen patients with a single lesion that grows very slowly for years. Stopping that drug at that time probably does a disservice to the patient. The same criteria are important for cytotoxic therapy in TKIs. You need to make sure that the patient is not symptomatic, that he or she is tolerating the drug well. Make sure that the life the patient is leading with the TKI is acceptable and appropriate for the patient. If all those criteria are met, only growth remains in an indicator lesion that you have determined is not symptomatic, and the patient is tolerating the drug well, the TKI should be continued and you should carefully reassess, reexamine, and reimage.

For patients in whom you decide to change therapy, it is very important to continue the TKI and/or substitute a drug that also effectively blocks EGFR signaling. We can only do that through a clinical trial here in the United States where we have only 1 EGFR-TKI available, but someday we will probably have more than 1 available and switching to another TKI may be a strategy. For today, you need to continue the erlotinib.

If you decide that a patient needs to be treated beyond the erlotinib for new symptoms or new lesions, do whatever local therapy is important -- for example, a brain metastasis would require some local care in many cases -- or add in a new drug to the TKI.

Unfortunately, like many issues in oncology, it is more complicated than just the word "progression." You need to very carefully see what progression means for the patient. You need to see what you know already about the biology of that patient's cancer through their initial treatment and make the best decision for the patient when progression is documented on an imaging study.

个人公众号:treeofhope
老马  博士一年级 发表于 2012-12-27 12:56:16 | 显示全部楼层 来自: 浙江温州

MET inhibition in lung cancer

本帖最后由 老马 于 2013-2-23 11:27 编辑

MET inhibition in lung cancer
http://www.tlcr.org/article/view/813/1406
Introduction Other SectionIntroduction Met discovery and mechanism of action Met pathway and cross-talks MET and NSCLC Met inhibitors Conclusions AcknowledgementsReferences

The discovery of new cancer-driver genes and the enforcement of molecules targeting them have changed the landscape of Non Small Cell Lung Cancer (NSCLC) treatment.

As a matter of fact, the previous scenario of advanced NSCLC treatment has been completely revolutionized, switching from a “one size fits all” approach to a personalized therapy.
Somatic mutations of the Epidermal Growth Factor Receptor (EGFR) tyrosine kinase domain positively correlated with clinical responsiveness to specific inhibitors: gefitinib, erlotinib and afatinib, two reversible and one irreversible EGFR inhibitors, have consistently demonstrated significant increase of Response Rate (RR) and Progression-Free-Survival (PFS) compared to standard chemotherapy in EGFR mutated NSCLC patients with advanced disease (1-7).
The Anaplastic Lymphoma Kinase (ALK), firstly identified from a chromosomal translocation leading to the production of merged proteins in Non-Hodgkin lymphomas, was then detected as a fusion with the echinoderm microtubule-associated protein-like 4 (EML4) in 6.7% of NSCLC patients (8,9). Crizotinib (PF02341066, Xalkori) targets EML4-ALK thus gaining astonishing response rates in a phase I/II trial and more recently in a phase III trial (10,11).
Unfortunately, other biomarkers already identified in NSCLC are still “undraggable” and one clear example is KRAS. KRAS is a member of the RAS family of oncogenesis, involved in signal transduction and tumorigenesis and its mutations, frequently in codons 12 and 13, have been reported in 20-30% NSCLCS (12-15). Some sign of activity came in the last year from a targeted agent (Selumetinib), which compared to standard chemotherapy in KRAS mutated patients gave interesting results in terms of RR and PFS (16).
Several other molecular markers’ alterations have been described in NSCLC such as: phosphatidylinositol 3-kinases (PI3K) (2%), lipid kinases that regenerate a key mediator between growth-factor receptors and intracellular downstream signaling pathways; ERBB-2 (2%); B-RAF (1-3%), a Ser-Thr kinase that links RAS GTPases to downstream proteins of the MAPK family, thus controlling cell proliferation; ROS1 (about 1%), oncogene that encodes a transmembrane tyrosine kinase receptor; AKT; RET and MET (17-21).
Since the first MET pathway description, several inhibitors have been preclinically and clinically tested, both alone and in combination with chemotherapy or other targeted therapies.

This paper will focus on MET biology, its role in the cell function and tumorigenesis, specifically in lung cancer, as well as on the molecules that target it.
--------------------------------------------------------------------------------

Met discovery and mechanism of action Other SectionIntroduction Met discovery and mechanism of action Met pathway and cross-talks MET and NSCLC Met inhibitors Conclusions AcknowledgementsReferences

Met is a heterodimer receptor tyrosine kinase composed of a α-chain and a β-chain, linked by a disulphide bond.
Met was originally isolated as the product of a human oncogene, trp-met, in tumor cells treated with a chemical carcinogen. Met gene encodes a 170-kD protein (p170met) that has constitutive and ligand-independent tyrosin-kinase activity. Met has pivotal functions in embryogenesis and organogenesis of placenta, liver, kidney, neurons and muscles (22-25).
Moreover, in vivo, Met receptor activation determines a phenomenon called “invasive growth”, which includes cell proliferation, scattering, survival, motility and invasion, epithelial-mesenchymal transition and branched morphogenesis (26,27).
The natural ligand for this receptor is the HGF, produced by stromal and mesenchymal cells, that acts primarily on Met-expressing epithelial cells in an endocrine and/or paracrine fashion (24,28). HGF-induced Met tyrosine kinase activation is regulated by paracrine ligand delivery, ligand activation at the target cell surface and ligand-activated receptor internalization and degradation (29). Going more into details, when HGF binds to the Met receptor, Met major autophosphorylation sites (located within the tyrosine kinase domain) are phosphorylated, with subsequent intrinsic catalytic activation of multiple signaling cascades involved in cell proliferation, survival, angiogenesis, morphogenesis, cell scattering, motility, migration and invasion. An activated docking site in the kinase domain further recruits intracellular adaptor molecules through the SH2 domains and other recognition motifs, such as GAB1 (a key coordinator of the cellular responses to Met). Downstream signaling of the GRB2-mitogen-activated protein kinase (MAPK) cascade, PI3K-mTOR pathway, and STAT pathway are eventually activated, mediating various cellular functions (27,30,31). Finally, in order to activate the receptor, proteolytic cleavage of proHGF is necessary (25).
HGF is mainly produced by stromal tissue like liver and bone marrow, and is expressed in a multitude of mesenchymal-derived cells. Being Met expression detected in the epithelium of most tissues, this indicates that HGF-Met signal transduction pathway contributes to mesenchymal-epithelial interactions (24,32-34).
Met downregulation occurs through rapid internalization of Met itself and subsequent degradation by the lysosome: this process is regulated by ligand-dependent ubiquitination of Met, a process also modulated by specific tyrosine phosphatases and recently identified as proteins decorin and LRIG1 (35,36).
Met can be altered through receptor overexpression, genomic amplification, mutations or alternative splicing. These alterations lead to signaling deregulation that can be mediated through ligand (HGF)-independent receptor activation or through its ligand (HGF)-dependent activation via autocrine (intratumoral HGF), paracrine (mesenchymal or microenvironmental HGF), or endocrine (circulatory HGF) loop signaling cascades (29).
HGF and Met are highly expressed in various stem and progenitors cells, but are only expressed as low levels in their mature cells (25). In preclinical animal models, whereas the overexpression of Met and/or HGF has been shown to stimulate tumorigenesis and metastasis, down-regulation of Met or HGF expression resulted in increased apoptosis and decreased tumor growth and blood vessel density (37-40). Moreover, Met interacts synergistically with VEGF to promote angiogenesis, cell proliferation and invasion (41). This occurs through the transcriptional up-regulation of the hypoxia inducible factor-1α and amplified HGF signaling, that resulted in both induction of invasion and increased expression of VEGF (41).
Met pathway is also one of the key players in the development of acquired resistance to VEGF pathway inhibitors: the inhibition of Met expression prevented hypoxia-induced invasion growth (42,43).
The increased Met expression described in case of response to ionizing radiation through the ATM-NFκB signaling pathway, could lead to radioresistance and cancer invasion (44).
--------------------------------------------------------------------------------

Met pathway and cross-talks Other SectionIntroduction Met discovery and mechanism of action Met pathway and cross-talks MET and NSCLC Met inhibitors Conclusions AcknowledgementsReferences

The cross-talk of Met with various signaling pathways is described in literature and that one between Met and EGFR/HER family receptors is particularly important in lung cancer (45-49).
Met and EGF family receptors are often described co-expressed in tumors and transactivation of Met depends on elevated expression of EGFR in many human tumors (46,50,51). Conversely, HGF stimulation promotes transactivation of EGFR in multiple cell lines, including NSCLC (49).
Cooperation between Met and EGFR occurs also indirectly: when Met activates Src, this lead to EGFR phosphorylation and the creation of docking sites for EGFR interactors involved in downstream signaling (52).

Moreover, through receptor cross-talk, Met exerts a key role in the development of resistance to EGFR family inhibitors. One example is the stimulation of HER-3 phosphorylation and signaling to Akt (a key signaling molecule required for cell survival and proliferation) when Met is amplified and overexpressed (53,54). Inhibition of Met in EGFR inhibitors resistant cells, either in vitro or in vivo, promotes apoptosis, tumor growth reduction and significant necrosis (49,53).

Met and EGFR inhibitors combined together, cooperatively abrogate ErbB3 signaling activation (49). An alternative mechanism in this context is the Src-induced EGFR phosphorylation (52).
Preclinical data also support that Met cross-talks and cooperates with other members of the EGF receptor family, including HER2, to enhance cell invasion and this lead to the possibility to explore therapeutic activity of dual Met and HER2 therapies (55,56).
Stimulation with both HGF and EGF enhances downstream activation of several signaling pathways including Akt, Erk and STAT3 in a way that Met inhibitors abolished their baseline phosphorylation (57,58).
The already mentioned interaction between decorin and LRIG1 proteins, promotes ligand-independent receptor downregulation and degradation of EGFR family members. Decorin binds to the EGFR family, inducing receptor dimerization, internalization and eventual lysosomal degradation, whereas LRIG1 and EGFR associated via their extracellular domains, allow enhanced EGFR phosphorylation. Thus, Met promotes resistance to VEGFR and EGFR inhibitors (59,60).
Cross-talk between Met and KRAS signaling has also been described both in preclinical and clinical findings (61,62). Met activates RAS directly or via a protein-tyrosine phosphatase (63). Similarly, PI3K could be directly activated by Met or indirectly by RAS protein (30).
Moreover, Met directly binds to and sequesters the Fas receptor. This interaction prevents Fas self-aggregation and ligand binding, thus inhibiting Fas activation and apoptosis (64).
Finally, preclinical studies exploring a combination of anti-Met therapeutic agents with mTOR inhibitors have also demonstrated an increased growth suppression, compared to mTOR inhibitors alone (62).
Met plays also a functional role in signaling pathways mediated by other membrane proteins. Integrin-dependent signaling could trigger ligand-independent Met phosphorylation following cellular adhesion, and Met and integrins might have independent yet synergistic roles in cell invasion. Plexins, single-pass transmembrane receptors for semaphorins, acts cooperatively with Met for cell adhesion and migration (45).
--------------------------------------------------------------------------------

MET and NSCLC Other SectionIntroduction Met discovery and mechanism of action Met pathway and cross-talks MET and NSCLC Met inhibitors Conclusions AcknowledgementsReferences

Met receptor is overexpressed in both Small Cell Lung Cancer (SCLC) and NSCLC, mainly in non-squamous histotype (65-67).
Recent tumor microarray expression analysis demonstrated a 72% Met expression in human lung cancer tissue and 40% Met receptor over-expression; such values are higher than in breast (16%) and ovarian cancer (31%), but lower than in renal (70%) and colorectal cancers (CRC; 78%) (67). Phospho-Met expression is found to be at the highest levels in lung cancer (73%), followed by ovarian (33%), breast (23%), and renal (18%) cancer (67).

Met gene amplification can guide the dependency of cell survival and proliferation upon the Met signaling, even in lung cancer cell lines. Blocking Met causes significant growth inhibition, G1-S arrest and apoptosis in cell lines harboring Met gene amplification. When Met is not amplified, its levels of activation are low and cells are unable to grow (68).

Different studies have reported primary Met amplification to be in the wide range of 2% to 21%, in NSCLC lung adenocarcinomas, particularly in TKI-naïve cohorts (69-72).

In lung cancer, Met receptor mutations were mainly found clustered in the non-tyrosine kinase domain, in the juxtamembrane (JM) domain and in the sema domain (67). These mutations are oncogenic activating variants, that result in a deletion in the juxtamembrane domain with enhanced oncogenic signaling, tumorigenicity, cell motility, and migration (27,73). Met kinase domain mutations have been found to be somatically selected in the metastatic tissues, compared with the primary solid cancers (74).
Literature data are quite discordant on the prognostic value of Met over-expression, amplification and mutation.
The overexpression of circulating Met in patients with NSCLC has been strongly associated with early tumor recurrence and patients with adenocarcinoma and Met amplification have also demonstrated a trend for poor prognosis (69,75,76).
Concerning the correlation between Met FISH status and clinical characteristics, only Okuda and colleagues demonstrated an association with male gender and smoking status, showing also a relationship with high Met gene copy number (77). In the same trial, both FISH positive and gene amplified cases had a worse prognosis, although the difference was not statistically significant and among the Met FISH-positive NSCLCs, patients with gene amplification showed not significantly worse OS compared to those with high polysomy.
All FISH-positive cases had squamous histology, adenocarcinoma had Met amplification: high Met gene copy number tended to have shorter OS and PFS than those with low Met gene copy number, being this difference statistically significant only in the squamous histotype.
Moreover, at multivariate analysis done on squamous histology, increased Met gene copy number and Met amplification were confirmed to be independent poor prognostic factors.
No significant difference in prognosis was found in patients having adenocarcinoma regardless Met FISH status in the korean study. In contrast, Beau-Faller and colleagues found a tendency toward shorter event-free survival in adenocarcinoma patients with increased Met gene copy number, whereas Kanteti and colleagues demonstrated that the high Met gene copy number in adenocarcinoma was associated with a trend of better prognosis (69). However, the above mentioned study has some critical methodology aspects as it was conducted on a small sample size and qPCR was used as test and not FISH, done on DNA samples extracted from formalin-fixed paraffin-embedded (FFPE) archival tumor tissues (70).
Capuzzo and colleagues found no patient with EGFR mutation was Met FISH positive, but increased Met gene copy number significantly correlated with EGFR FISH-positive status (78).
Acquired Met amplification has also been linked to approximately 22% of non-T790M mediated secondary gefitinib resistance in NSCLC patients, although it can also occur concurrently but independently (52,53,78-80).
Using in vitro cell line models, the Met gene amplification in gefitinib-resistant cell clones was identified (53).
Rho and colleagues tried to demonstrate that Met activation, rather than gene amplification, is sufficient to promote EGFR resistance, but the activation appear to be secondary to increase passage numbers rather than to EGFR-Tki exposure (81).
More recently, two prospective analyses have investigated the mechanism of EGFR-Tki resistance through the tissue rebiopsy: high Met gene copy number was found in 11% and 5% of the tissue samples, respectively (82,83).
--------------------------------------------------------------------------------

Met inhibitors Other SectionIntroduction Met discovery and mechanism of action Met pathway and cross-talks MET and NSCLC Met inhibitors Conclusions AcknowledgementsReferences

Several inhibitors have been tested so far: they can be classified according to their mechanism of action in selective Met inhibitors, unselective Met inhibitors and antibodies targeting Met or HGF (Figure 1, Table 1).

Table 1 Ongoing trials on Met inhibitors
Full table  Figure 1. Met inhibitors in the clinic Selective Met inhibitors
Tivantinib
Tivantinib (ARQ 197) is the first non-ATP-competitive small molecule that selectively targets the Met RTK, locking and stabilizing the kinase in a “closed” and “inactive” conformation, causing the disruption of Met phosphorylation and the downstream signaling.

Moreover, tivantinib enhances Met degradation through the ubiquitin/proteasome pathway in vitro, induces apoptosis in Met activated cell-lines and it’s active in multiple human cancer xenografts (84,85).

Tivantinib acts synergically with antiangiogenentc drugs in preclinical studies on solid tumor cell lines (86).

Studies in vitro and in vivo demonstrated its activity in several types of cancer such as breast, colorectal and gastric cancer (85,87).

Met cancer expressing cell lines treated with tivantinib displayed either a dose-dependent loss of proliferative capacity or caspase-dependent Met apoptosis, which positively correlated with either ligand-dependent Met activity or constitutively active Met. Tivantinib does not exert any activity in cancer cell lines not expressing Met or phospho-Met.

Tivantinib has been investigated in three phase I trials, as single agent and in combination.

In the first dose-escalation phase I trial, tivantinib is administered as single-agent in patients with advanced solid tumors. Initially, an intermittent dosing was planned but, due to the bradycardia experienced in the other phase I trial using this schedule, the protocol was amended and the following 79 patients received a continuous dose (88).

No MTD was reached in this study and less than 33% of patients experienced DLTs at any given dose. Thus, the recommended phase II dose was confirmed at 360 mg twice a day as per a concomitant phase I study, where this MTD was identified (88).

The most commonly reported drug-related adverse events of any grade included fatigue, gastrointestinal (GI) disorders (nausea, vomiting and diarrhea) and anemia.

Pharmacokinetic was linear. There was considerable inter-patient variability, but no relationship between drug-related adverse events (AEs), dose and extent of tivantinib exposure; consequently, this inter-patient variability was not considered relevant for its clinical safety. Partial responses registered in this trial were equal to 4.8% (89).

In another phase I trial, two formulations of tivantinib were tested: the amorphous and the crystalline A formulation. The trial was lead in a single institution, the Royal Marsden Hospital (Sutton, United Kingdom) and highlighted the following DLTs: one patient had grade 3 fatigue at 200 mg, one patient presented a grade 3 febrile neutropenia, one other a grade 3 mucositis, one a grade 3 palmar-plantar erythrodysesthesia and one a grade 3 hypokalemia at 400 mg. The MTDs- recommended phase 2 doses (RP2Ds) were 300 mg bi-daily for the amorphous formulation and 360 mg bi-daily for the crystalline A formulation. The main grade 1-2 AEs, all generally self-limiting, were fatigue (15.7%), nausea (13.7%), vomiting (11.8%). Tivantinib is metabolized by CYP2C19: one patient with CYP2C19 deficiency experienced grade 4 febrile neutropenia and grade 3 mucositis as the drug’s AUC was 3-fold higher (90).

The crystalline A formulation of tivantinib resulted in lower drug exposure at 300 and 360 mg twice daily, compared with the amorphous form at 300 mg twice daily (likely due to different dissolution characteristics). RECIST stable disease ≥4 months was the best response in 14 patients, together with minor tumor regressions (88).

As the ratio of the poor metabolizers of CYP2C19 in Asians is around 20% (while is very low in Caucasians), a Japanese phase I trial was designed to evaluate drug’s safety profile of tivantinib in this group of patients with metastatic solid tumors and the drug was well tolerated, but CYP2C19 genotype clearly affected the exposure and the RP2Ds differed for “no poor metabolizers” (360 mg bi-daily) and for “poor metabolizers” (240 mg bi-daily). Most common AEs were similar to those mentioned above (91). A phase III trial was conducted in Asia in advanced NSCLC patients, comparing erlotinib + tivantinib versus erlotinib + placebo at the dose calculated considering the CYP2C19 polymorphism (92). A press release in August 2012 announced a suspension in the accrual for this study, due to suspected cases of interstitial lung disease (93).

Based on the preclinical data showing a synergistic action between EGFR-TKi and Met inhibitors, an open-label sequential dose escalation phase I trial on tivantinib + erlotinib was set up. Thirty-two metastatic cancer patients were included: 59% were males, 75% PS 1 and mean age was 60 years. The MTD was not established, however, the RP2D was 360 mg bi-daily for tivantinib and 150 mg daily for erlotinib. Two DLT were experienced at 360 mg (grade 4 neutropenia, grade 3 thrombocytopenia), none at 240 or 120 mg. The most common AEs were cutaneous rash, fatigue, nausea, abdominal pain, diarrhea, bradycardia and anemia, mostly grade 1 and 2. No drug related death, but 11% grade 3-4 neutropenia and 8% grade 3-4 nausea were recorded (94).

This combination of erlotinib (150 mg daily) + tivantinib (360 mg bi-daily) every 4 weeks was further studied in a phase II, double-blind, randomized open-label study in comparison with erlotinib 150 mg daily + placebo, in previously treated locally advanced or metastatic NSCLC patients. One hundred and sixty-seven patients were enrolled and homogeneously distributed between the two arms (mainly males, never or former smoker, with stage IV disease and adenocarcinoma histology): 10% in the combination arm versus 18% in the standard arm presented an EGFR mutation, 10% versus 17% a KRAS mutation, 26% versus 26.5% had 4 or more MET gene copy number. The ORR was 10% for erlotinib + tivantinib versus 7% for the control arm.

Median investigator’s PFS was 3.8 months for the tivantinib + erlotinib arm versus 2.3 months for the erlotinib + placebo arm (HR=0.81, P=0.24); the reviewer’s PFS was 3.6 versus 2 months (HR=0.74, P=0.09). Median OS was 8.5 for the investigational arm versus 6.9 months for the control arm (HR=0.87, P=0.47). Pre-planned exploratory survival analysis in non-squamous histology showed a trend of benefit from the combination arm in both PFS (HR=0.71) and OS (HR=0.72). Even in a small number of patients, the subgroup analysis showed an advantage in terms of PFS for EGFR wild type (HR=0.70), KRAS mutated patients (HR=0.76) and for Met FISH positive patients (>5, HR=0.45).

Treatment was well tolerated both in the investigational and in the control arm: low grade rash (9.5% versus 7.2%) and diarrhea (7.1% versus 7.2%), fatigue (4.8% versus 6%), nausea (1.2% versus 4.8%), vomiting (3.6% versus 1.2%), dyspnea (7.1% versus 13.3%), anemia (6% versus 7.2%) were the most common reported toxicities (95).

On the basis of data coming from this phase II trial, the phase III MARQUEE trial was designed in non-squamous NSCLC patients with the same schema, having the overall survival (OS) as primary end-point. Unfortunately, a press release in October 2012, revealed that the primary end point in the intent to treat population was not met, but no further data are yet available (96,97).

Others selective Met inhibitors
PF-04217903 is a selective ATP-competitive small inhibitor of Met kinase. It inhibits tumor cell proliferation, survival, migration/invasion in Met-amplified cell lines in vitro, and shows marked antitumor activity in tumor models harbouring either Met gene amplification or a HGF/Met autocrine loop. PF-04217903 also demonstrates potent antiangiogenic properties in vitro and in vivo (98). In 2012 a phase I trial with PF-04217903 in patients with advanced solid tumors was prematurely discontinued, due to strategic development decision by Pfizer. No safety concerns were reported (99).

AMG 337 is a selective inhibitor of the proto-oncogene Met thereby disrupting Met signal transduction pathway. A phase I, open-label, sequential dose escalation and expansion study with AMG 337 in subjects with advanced solid tumors is currently ongoing (100) (Table 1).

INCB028060 is an oral potent and highly selective Met inhibitor, capable of suppressing tumor growth in vivo at doses that are extremely well tolerated (101,102).

Good tolerance was confirmed in a phase I standard 3+3 dose-escalation study once or twice daily on a continuous 28-day schedule in patients with advanced solid tumors. The MTD was not reached and no grade 3-4 AEs were noted, except grade 3 ALT increase in a patient with liver metastases and grade 2 ALT levels at baseline. Grade 1-2 AEs experienced were mild tremor, fatigue, nausea, diarrhea, indigestion and headache (103).

Non-selective Met inhibitors
Crizotinib
Crizotinib was synthesized primarily as a Met inhibitor. It was engineered based on interactions of a precursor (PHA-665752) with the ATP-binding sites of the Met kinase domain thus resulting in displacement of the kinase activation loop, that interferes with ATP and substrate binding to the Met receptor tyrosine kinase. Crizotinib was designed in order to be less lipophilic and to have a small hinge binder with the possibility to better interact in the kinase pocket (104).

Crizotinib was proved to be active in NSCLC cell lines carrying Met amplification. However, no activity was described in Met mutated, EGFR mutated or normal cell lines. Moreover, crizotinib markedly inhibited AKT, Met and ERK phosphorylation. By doing that, it induced apoptosis even though a mediation of BIM up-regulation (pro-apoptotic member of the Bcl-2 family) and survivin down-regulation (a member of the inhibitor of apoptosis protein family) has also been reported.

Interestingly, in Met or EGFR mutated but also in normal cell lines, whit a low Met phosphorylation, the Met phosphorylation is completely inhibited, whereas the ERK and AKT are not (105).

During drug development, Ou and colleagues described a case of prolonged partial response to crizotinib in a NSCLC patient carrying Met amplification (defined as Met/CEP7 ratio >5) but no ALK translocation (106).

The first phase I trial was designed as open-label, multicenter, to evaluate safety and efficacy of crizotinib: this study was emended with an expanded cohort for patients with lung cancers carrying ALK rearrangements. The recommended crizotinib dose was 250 mg twice daily in 28-day cycles.

In the overall NSCLC population a phase I trial investigated crizotinib in association to dose escalating erlotinib: 5 DLTs were reported (at 150/100 mg grade 2 vomiting, grade 2 esophagitis and dysphagia, grade 3 diarrhea and dehydration; at 200/100 mg, grade 3 dry eye and grade 3 esophagitis). Ninety-two percent of the patients experienced treatment-related AEs, mainly grade 1 or grade 2: diarrhea (72%), rash (56%) and fatigue (44%) (107).

Another phase I trials evaluated crizotinib in combination with dacomitinib, an irreversible pan-erb inhibitor in previously treated advanced NSCLC patients (108).

Cabozantinib
Cabozantinib (XL184) is a potent Met/VEGFR2/RET/KIT/AXL/FLT3 inhibitor that targets tumor survival, metastasization and angiogenesis.

It selectively inhibits KIT, RET, AXL, TIE2 and FLT3 (all kinases implicated in tumor pathobiology) through strong, reversible, ATP-competitive binding. Moreover, cabozantinib inhibits HGF and VEGF-mediated cell migration and also Met and VEGFR phosphorylation and the tubule formation, with no evidence of cytotoxicity.

This effect described in vitro, turned into in vivo significant tumor regression, without any relevant toxicity (109).

Several phase I trials have already been published verifying the schedule, the formulation, the dose of the drug, both as single-agent and in combination with other molecules.

Kurzrock and colleagues studied single-agent cabozantinib both in suspension and capsule formulation, at intermittent (5 days on, 9 off) and continuous schedule: MTD was defined at 175 mg continuous schedule, being DLT mucositis, elevated lipase and altered liver function (110).

The continuous dose was further investigated in a Japanese only population: MTD was 60 mg, being grade 3 hypertension the DLT (111).

Regarding combination regimens, a phase I study analyzed the interaction of the combination cabozantinib and rosiglitazone, as the latter is a CYP2C8 substrate, but no interaction was found between these two compounds (112,113).

Cabozantinib was further studied in several phase II trials in different tumor types. Among them, one phase II trial investigated treatment with cabozantinib in NSCLC patients previously treated with anti-EGFR TKi (50%) and anti-VEGF therapies (32%). At week 12 the ORR was 10% and the overall DCR 40%. No difference in terms of PFS (median 4.2 months) was seen in the two populations according to the treatment response at 12 weeks. The most common grade 3-4 events were diarrhea (7%), palmar-plantar erythrodyesthesia (8%), fatigue (13%) and asthenia (7%) (114).

Likewise tivantinib, also cabozantinib was tested together with erlotinib or gefitinib in vivo and in vitro in EGFR TKi resistant NSCLC xenograft models harboring Met amplification. Gefitinib and cabozantinib were tested on gefitinib resistant cell lines either alone and in combination and the two molecules together were substantially more potent than each drug alone (>50% inhibition). The same result was obtained with the combination of erlotinib and cabozantinib on erlotinib resistant cell lines (115).

The combination of cabozantinib and erlotinib was tested on 54 NSCLC patients in a phase Ib/II study. Patients were divided into 5 cohorts in two parallel arms: arm A (75 mg cabozantinib + 100 mg erlotinib; 125 mg cabozantinib + 100 mg erlotinib; 125 mg cabozantinib + 50 mg erlotinib) and arm B (75 mg cabozantinib +150 mg erlotinib; 50 mg cabozantinib +150 mg erlotinib). Twelve patients experienced at least 1 DLT: diarrhea, increased AST levels, palmar-plantar erythrodysesthesia, mucositis, hypertension, hypokalemia, elevated lipase and fatigue. The most common grade 3-4 adverse events were diarrhea (26%), fatigue (15%), dyspnea (12%) and hypoxia (9%) (116).

In advanced NSCLC patients two phase II trials are ongoing: the first one randomizes EGFR wild type patients to erlotinib, cabozantinib or erlotinib plus cabozantinib as second or third line therapy; the second study investigates cabozantinib in patients with KIF5B/RET positive NSCLC (117,118) (Table 1).

Foretinib
Foretinib (XL-880, EXEL-2880) is an oral multi-kinase inhibitor developed to target Met and several other receptor tyrosine kinases involved in tumor angiogenesis. It is an ATP-competitive inhibitor and binds the ATP pocket of both Met and VEGFR-2 tyrosine kinase domains with high affinity.

Both in vitro and in vivo, foretinib inhibits Met and VEGF receptor-2 (VEGFR-2) and have high in vitro affinity for PDGFRb, Tie-2, RON, Kit, and FLT3 kinases, preventing tumor growth through a direct effect on tumor cell proliferation and inhibition of invasion and angiogenesis, mediated by HGF and VEGF receptor (119).

Two phase I trials have been published: the first investigated foretinib administered for 5 consecutive days every 14 days in a 3+3 dose escalation study; in the second study foretinib was administered once daily for 28 days. Both trials were conducted in patients with metastatic or unresectable solid tumors. MDT was defined as 3.6 mg/kg for 5 consecutive days every 14 days in the first study and as 80 mg daily in the second; DLTs in the first study included grade 3 elevations in aspartate aminotransferase and lipase, whereas in the second trial hypertension, dehydration and diarrhea were described.

Additional AEs in both studies included hypertension, fatigue, diarrhea, vomiting, proteinuria, and hematuria. In these studies no responses were observed and most of patients achieved a stable disease as best response (120,121).

A phase I, open-label, randomized, 2-part crossover study assessed the safety, pharmacokinetics and relative bioavailability of single doses of foretinib free base tablet formulation compared to a bisphosphate salt capsule formulation: both were well tolerated and their pharmacokinetics and relative bioavailability were not clinically different (122).

On the basis of preclinical data, showing that combining foretinib with erlotinib or lapatinib effectively decrease the phosphorylation of Met, HER1, HER2, HER3, AKT, and ERK in cell lines, a phase I/II study of erlotinib in association or not with foretinib in previously treated NSCLC patients has been designed and is currently ongoing (123,124) (Table 1).

Golvatinib
Golvatinib (E7050) is a novel small molecule ATP-competitive inhibitor of Met receptor, that potently and selectively inhibits the autophosphorylation of Met and VEGF-induced phosphorylation of VEGFR (125).

Golvatinib also circumvents resistance to reversible, irreversible, and mutant-selective EGFR-TKIs induced by exogenous and/or endogenous HGF in EGFR mutant lung cancer cell lines, by blocking the Met/Gab1/PI3K/Akt pathway in vitro and also prevents the emergence of gefitinib-resistant cells, induced by continuous exposure to HGF (126).

A phase I study with oral daily golvatinib administered continuously once a day in patients with advanced solid tumors was performed. Three DLTs were observed: grade 3 increase in GGT and alkaline phosphatase levels and grade 3 fatigue, all at 450 mg. The MTD was determined to be 400 mg every day. Frequently occurring AEs were fatigue (68%), diarrhea (65%), nausea (62%), vomiting (53%), decreased appetite (47%), ALT increase (38%) and AST increase (23%). No grade 4 AEs were observed (127).

Other molecules
MGC D265 is an oral receptor tyrosine kinase inhibitor targeting Met, VEGF, RON and Tie2. Preclinical data have demonstrated synergism of action with erlotinib and early clinical trials are currently ongoing (128) (Table 1).

ANG707 is another non-selective Met inhibitor under investigation in early phase trials (129).

Antibodies
Antibodies against Met
Onartuzumab (MetMab)
MetMAb is a recombinant, fully humanized, monovalent monoclonal anti-Met antibody based on the human IgG1k framework sequence. It binds in the sema domain of Met within the extracellular domain, where it acts to inhibit HGF binding and initiation of receptor activation. The unique monovalent design of MetMAb eliminates the potential for Met activation via antibody-driven receptor dimerization (130).

The activity shown in vitro by MetMAb did not translate into a full activity in vivo: only about 65% tumor inhibition was demonstrated, indicating that blockade of HGF by MetMAb is not sufficient for full tumor inhibition in specific tumors (130).

A phase I trial investigated sequential 3+3 dose-escalation of endovenous MetMAb in advanced solid tumors: MetMAb was three weekly intravenously administered, both as single agent and in combination with bevacizumab 15 mg/kg every three weeks, until progression.

Most frequent MetMAb AEs as single-agent were: fatigue (56%), peripheral edema (35%), decreased appetite (32%), constipation (29%), nausea (27%), vomiting (24%) and hypoalbuminemia (24%); there was no consistent relationship between AEs and dose level.

Grade 3 AEs were peripheral edema (9%), abdominal pain, AST increase, fever and hyponatremia. No Grade 4 toxicity was observed. The combination arm had similar toxicities; no grade 3 or 4 toxicity was experienced. MTD was not reached. The best response was stable disease (131).

The phase II trial was a global, randomized, double-blind trial evaluating the combination of MetMAb + erlotinib versus placebo + erlotinib in second/third line NSCLC advanced patients. One hundred and twenty-eight NSCLC patients were enrolled with a baseline immunohistochemical evaluation of Met: 54% of the patients were considered as Met positive (high protein expression at IHC). Met positive patients treated in the experimental arm had a significantly higher PFS (3.0 vs. 1.5 months; HR 0.47; P=0.01) and OS (12.6 vs. 4.6 months; HR 0.37; P=0.002) (132).

Based on phase II data, a randomized, phase III, multicenter, double-blind, placebo-controlled study evaluating the efficacy and safety of onartuzumab in combination with erlotinib in patients with Met positive NSCLC who have received standard chemotherapy for advanced disease is currently recruiting patients (133) (Table 1).

The positive results of the phase I trial on MetMAb in combination with bevacizumab have paved the way to the ongoing randomized phase II multicentric double-blind placebo-controlled study evaluating the efficacy and safety of MetMAb in combination with either bevacizumab + platinum + paclitaxel or pemetrexed + platinum as first-line treatment in patients with stage IIIB and IV non-squamous NSCLC (134).

Antibodies against HGF
Ficlatuzumab
Ficlatuzumab (AV-299) is a potent hepatocyte growth factor (HGF) inhibitor IgG1 monoclonal antibody, that binds to the HGF ligand with high affinity and specificity. Ficlatuzumab was studied in two phase I trials and one phase II study. In both phase I trials it was associated with gefitinib and erlotinib. In the first phase I trial ficlatuzumab was biweekly administered intravenously over 30-60 minutes both as single-agent and in combination with erlotinib at 150 mg continuously in advanced solid tumors. There were no DLT in the monotherapy arm; consequently no MTD was identified.

For the combination arm there was one DLT (grade 3 mucositis). The RP2D for both monotherapy and combination regimen was defined as 20 mg/kg every 2 weeks. Ficlatuzumab as a single-agent demonstrated a stabilisation of disease in 50% of the cases (135).

The second phase Ib trial enrolled only Asiatic patients with unresectable NSCLC: ficlatuzumab was administered intravenously every 2 weeks at two dose levels (10 and 20 mg/kg) in combination with gefitinib at 250 mg daily. No DLTs were observed in the dose-escalation cohorts; 20 mg/kg of ficlatuzumab every 2 weeks + gefitinib 250 mg daily was selected as RP2D. Among 12 patients in the 20 mg/kg cohort, 5 partial responses were achieved (136). Most frequent treatment-emergent adverse events (AEs) were fatigue (27-33%), dermatitis acneiform (53%, particularly for the combination regimens), diarrhea (33-46%) and edema (16-27%) for both single-agent and combination therapy (135,136).

The efficacy of ficlatuzumab together with gefitinib was further investigated in a multicenter, open-label, exploratory, 2-arm randomized phase 2 study in previously untreated Asian NSCLC patients with the doses defined in the phase I. One-hundred eighty-eight patients were randomized with a baseline evaluation of Met by IHC and gene copy number. In the low Met group, ORR (41 versus 22%) and median PFS (7.3 versus 2.8 m) favored the combination regimen with a manageable toxicity profile (137).

Rilotumumab
Rilotumumab (AMG 102) is a fully human monoclonal antibody that selectively targets and neutralizes hepatocyte growth factor/scatter factor (HGF/SF). It preferentially bound to the β-chain of the human, mature, active form of HGF, and had no apparent effect on proteolytic processing of the inactive HGF precursor (138).

Two phase I trials have been published so far with AMG 102 in advanced refractory solid tumors: one as single agent and one in combination with bevacizumab or motesanib (139).

In the monotherapy trial, AMG 102 was well tolerated up to the planned maximum dose of 20 mg/kg, MTD was not reached and pharmacokinetic was linear. Two patients experienced DLTs: one grade 3 hypoxia and grade 3 dyspnea (0.5 mg/kg cohort) and one grade 3 upper GI hemorrhage (1 mg/kg cohort). Treatment-related AEs were generally mild and included fatigue (13%), constipation (8%), nausea (8%), vomiting (5%), anorexia (5%), myalgia (5%), and hypertension (5%). Seventy percent of the evaluable patients had a SD as best response (139).

The phase Ib combination study sequentially enrolled patients into four cohorts, but the number of those receiving AMG 102 plus motesanib was insufficient to adequately assess safety and the accrual was early suspended because of reports of cholecystitis in other motesanib studies. No dose-limiting toxicities were reported and the combination of AMG 102 with bevacizumab seemed to have acceptable toxicity. AEs were generally mild and included fatigue (75%), nausea (58%), constipation (42%) and peripheral edema (42%) (140).

TAK 701
TAK-701 is a humanized monoclonal antibody that binds HGF thus inhibiting its bound to Met receptor. TAK-701 in combination with gefitinib blocks the phosphorylation of Met, EGFR, extracellular signal-regulated kinase, and AKT in HGF expressing human NSCLC cell lines with an activating EGFR mutation. Combination therapy also markedly inhibited the tumor growth in vivo (141).

Preliminary data of a phase I study in advanced solid malignancies with TAK-701 showed that the most common AEs were cough, abdominal pain, constipation and fatigue, all grade 1-2. There were 3 grade 3 AEs (gastrointestinal ileus, pleural effusion, urinary tract infection) and 1 grade 4 AE (dyspnea). No DLT was found and the MTD has not been reached (142).
--------------------------------------------------------------------------------
Conclusions Other SectionIntroduction Met discovery and mechanism of action Met pathway and cross-talks MET and NSCLC Met inhibitors Conclusions AcknowledgementsReferences
In patients with advanced NSCLC, a correct definition of the histotype is still the first step to design a proper therapeutic algorithm, but personalized molecular diagnosis is becoming more and more relevant.
Genetically defined subsets of cancers may share dependence on a specific signaling pathway: specific inhibitors targeting these pathways would be most effectively tested in patient populations characterized by molecular markers.
Moreover, genetic events that arise and are selected during tumor progression may become essential for tumor survival, a phenomenon generally described as “oncogene addiction”: cancer cells appear to depend on a single overactive oncogene to proliferate and survive (143). Optimal case selection, diagnostic and pharmacodynamic biomarker development, the identification and testing of rationally designed anticancer drugs and combination strategies are crucial to develop the best treatment for the right patient (144).
New generations of molecularly targeted drugs will allow more personalized medicine and more efficacious and less toxic antitumor therapies in patients with defined molecular aberrations, sparing normal cells thus sparing toxicity (145,146).
Met can act as an ‘oncogene expedient’ even in absence of genetic alterations and might potentiate the effect of other oncogenes, promote malignant progression and participate in tumor angiogenesis (147).
Met dysregulation correlates with disease prognosis in numerous cancers and represents a possible target for personalized treatment. The clinical efficacy of Met targeting agents in lung cancer needs further details from the ongoing trials as well as more information are necessary to establish the most appropriate diagnostic test to identify Met expression or amplification.
Several molecules are currently under investigation and two of them already reached phase III trials in advanced NSCLC.
个人公众号:treeofhope
老马  博士一年级 发表于 2012-12-27 12:56:19 | 显示全部楼层 来自: 浙江温州
本帖最后由 老马 于 2013-5-30 01:49 编辑

T790M and acquired resistance of EGFR TKI: a literature review of clinical reports
http://www.jthoracdis.com/article/view/64/127
Comparison of T790M detection methods
In most cases, DS plus PCR were used to detect EGFR mutation status including T790M. Generally, PCR, especially mutant-enriched PCR is a more sensitive and highly specific assay that can detect one copy of mutant allele among as many as 103 to 104 copies of wild-type alleles when compared to direct DNA sequencing. The use of this assay has been validated with various clinical specimens, including biopsy and pleural fluid specimens .
The use of Scorpion Amplification Refractory Mutation System (SARM) was also reported, in which the two technologies ARMS and Scorpion were combined to detect T790M in real-time PCR reactions . Compared to traditional PCR, SARM system was more sensitive.
In one clinical study, both SARM and WAVE/Surveyor methods were employed for the detection of EGFR activation and resistant mutations using plasma DNA from TKI resistant patients. Eight out of nine T790M positive patients were detected by SARM, while WAVE-Surveyor system detected four, and three were detected by both methods. Whole genome amplification of samples seems to have the greatest effect on the detection of T790M. Furthermore, whole genome amplification of DNA samples was also investigated to determine if the detection of additional EGFR mutations would be facilitated. For EGFR del E746_A750 and L858R, whole genome amplification identified only four additional patients with mutations, whereas for EGFR T790, whole genome amplification resulted in the identification of 10 additional patients (P = 0.011). This may be explained by the fact that T790M often exists as a rare allele and thus may go undetected in the absence of whole genome amplification. The results suggest that whole genome amplification should be incorporated in future noninvasive method in order to aid in monitoring drug resistance and in directing the course of subsequent therapy.
In general, PCR, especially mutant PCR is more sensitive than direct sequencing. Since T790M is often present as a minor allele, whole genome amplification could improve detection of T790M.
To sum up, acquired-resistant patients usually undergo a dramatic response or partial response for around 10 months before they develop resistance to TKI. Few patients among them can be detected T790M positive; but after the resistance developed, T790M mutations account for half of these cases. Meanwhile, in our analysis, EGFR T790M always coexist with other resistance mutations such as MET amplification. The switch therapy from gefitinib to erlotinib might be useful for those originally well-responded patients. Irreversible TKIs in development might also show promises in overcoming T790M-induced resistance. The use of high-sensitivity analytical techniques may aid in finding suitable individualized therapy for patients based on their mutational status.


At present, noninvasive genotyping should not replace the gold standard of repeated tumor biopsy, because there are potential limitations to all plasma-based tumor DNA genotyping methods. Firstly, the technique lacks the ability to determine whether the isolated DNA is truly tumor-derived. One potential method to overcome this limitation is to specifically isolate circulating tumor cells and use them for the genotyping studies. Secondly, it would be difficult to determine whether the plasma-derived DNA changes are truly reflective of the genomic changes of the bulk of the cancer. It will be important to correlate the plasma-based studies with tumor-based studies. An additional factor that needs to be considered in noninvasive evaluation of EGFR T790M is the time between the development of resistance and the collection of the plasma DNA specimen for evaluation. Validation of the noninvasive method in prospective clinical trials is necessary to determine its sensitivity and specificity (1). The success of this method has the potential to eliminate the need for repeated tumor biopsies in the detection of T790M mutation. In the future, the incorporation of more sensitive systems, such as those based on high-performance liquid chromatography (HPLC), tandem mass spectrometry or high-density picoliter reactors may also be useful for detecting EGFR T790M.
个人公众号:treeofhope
老马  博士一年级 发表于 2012-12-27 12:56:25 | 显示全部楼层 来自: 浙江温州
本帖最后由 老马 于 2013-5-30 02:31 编辑

De novo EGFR T790M mutation modifies outcome to second-line erlotinib in non-small cell lung cancer (NSCLC) according to metastatic site and upfront chemotherapy
http://cancerres.aacrjournals.or ... etingAbstracts/4112
Rafael Rosell1, Miguel Angel Molina2, Jose Javier Sanchez3, Miquel Taron1, Susana Benlloch2, Teresa Moran1, Enric Carcereny1, Felipe Cardenal4, Bartomeu Massuti5, and Ignacio Magri2
1Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
2Pangaea Biotech, Dexeus University Institute, Barcelona, Spain
3Autonomous University of Madrid, Madrid, Spain
4Catalan Institute of Oncology, Hospital Duran i Reynals, Hospitalet de Llobregat, Barcelona, Spain
5Hospital General de Alicante, Alicante, Spain.

EGFR T790M mutation is associated with shorter progression-free survival (PFS) (12 months [m] vs 18 m; P=0.02). We hypothesized that the site of metastases (mets) and/or prior chemotherapy could also influence outcome in these p.

Methods: The T790M mutation was assessed in 129 advanced NSCLC p by TaqMan assay in the presence of a peptide-nucleic acid designed to inhibit the amplification of the wild-type allele.

Results: De novo T790M mutations were identified in 35% (45 of 129) of EGFR-mutant p before receiving erlotinib. The T790M mutation was detected more frequently in p with bone mets (35.6% vs 16.7%; P=0.03). PFS was 20 m for 58 p with a deletion in EGFR exon 19 (del 19) but without the T790M mutation vs 12 m for 23 p with both del 19 and the T790M mutation (P=0.03), but with no difference in MS between these two groups (31 m vs 29 m; P=0.56). PFS was 15 m for 26 p with the L858R mutation but without the T790M mutation vs 16 m for 22 p with both the L858R and T790M mutations (P=0.83), with no difference in MS between these two groups (27 m vs 21 m; P=0.81). When p with T790M were divided according to the presence of brain mets, PFS was 1 m for 4 p with brain mets vs 13 m for 41 p without brain mets (P=0.002), while MS was 6 m for p with brain mets vs 36 m for those without (P=0.009). No effect on PFS or MS was observed in p with the T790M mutation according to lung, liver, pleura or bone mets. In the multivariate analysis, the presence of the T790M mutation did not increase the risk of short MS (HR, 1.3; P=0.49), while having received prior chemotherapy was associated with longer MS (HR, 0.48; P=0.02).

Conclusions: The de novo T790M mutation is a marker for poor prognosis in p with brain mets. Upfront chemotherapy can play a role in the management of NSCLC p with the de novo T790M mutation.

个人公众号:treeofhope
老马  博士一年级 发表于 2012-12-27 12:56:28 | 显示全部楼层 来自: 浙江温州
本帖最后由 老马 于 2013-5-30 09:04 编辑

T790M的起源、遗传易感性和体内自然选择
作者:余细勇:陈思远    作者单位:1. 广东省人民医院 a.医学研究中心; b. 肿瘤中心; 2. 广东省肺癌研究所, 广州 510080
http://www.cnki.com.cn/Article/CJFDTotal-YEBM200804006.htm
1 文献来源

    ①Bell WD, Gore I, Okimoto AR, et al. Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR[J]. Nat Gene, 2005,37(12):1315-1316.

    ② Balak NM, Gong Y, Riely JG, et al. Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor-mutant lung adeno-carcinomas with acquired resistance to kinaseinhibitors[J]. Clin Cancer Res, 2006,12(21):6494-6501.

    ③Jackman DM, Holmes AJ, Lindeman N, et al. Response and resistance in a non-small-cell lung cancer patient with an epidermal growth factor receptor mutation and leptomeningeal metastases treated with high-dose Gefitinib[J]. J Clin Oncol, 2006,24(27):4517-4520.

    2 证据水平

    2b。

    3 背 景

    研究一: 酪氨酸激酶抑制剂(tyrosine kinase inhibitors, TKI)在部分非小细胞肺癌(non-small cell lung cancer, NSCLC)患者,尤其是非吸烟者、妇女和患腺癌伴有支气管肺泡分化者中取得了较好的疗效。以往研究认为,表皮生长因子受体(epidermal growth factor receptor,EGFR)T790M突变是在应用TKI治疗后出现的继发突变。然而在另一些研究中发现,从未给予TKI治疗的NSCLC患者中,也可发现T790M突变。

    研究二: 肺腺癌患者应用TKI治疗后可继发EGFR T790M突变,而T790M突变常见于TKI治疗获得性耐药患者。

    研究三:晚期NSCLC伴脑转移病人应用常规剂量的吉非替尼治疗常达不到理想的疗效。

    4 目 的

    探讨EGFR基因20外显子T790M突变的起源、遗传易感性及体内自然选择过程。

    5 研究设计

    条件:

    研究一: Massachusetts General Hospital Cancer Center and Department of Pathology, Harvard Medical School和Bruno Cancer Center,St. Vincent’s Hospital。

    研究二: Department of Medicine, Weill Medical College of Cornell University。

    研究三: Department of Medical Oncology, Dana-Farber Cancer Institute; Department of Me-dicine, Brigham and Women’s Hospital; Harvard Medical School。

    研究方法:

    研究一: 反转录PCR,基因测序,家系分析等。

    研究二: 限制性片段多态性分析(restriction fragment length polymorphism,RFLP),反转录PCR,基因测序,免疫印迹和细胞生长抑制分析等。

    研究三: 腰椎穿刺脑脊液分析,血清转氨酶测定,CT、MRI等影像学分析,体外肿瘤细胞药物抑制分析,基因测序,变性高效液相色谱(denaturing high-performance liquid chromatography,DHPLC)检测。

    研究对象:

    研究一:3代8人中6位患肺泡细胞癌的欧洲家庭,肿瘤样本来源为Ⅲ-1手术切除的左肺5个独立原发病灶,Ⅲ-2肺癌转移病灶,Ⅲ-3和Ⅲ-4外周血单核细胞,其中Ⅲ-1为TKI治疗有效,Ⅲ-2为TKI治疗耐药。

    研究二: 16位接受TKI治疗的肺腺癌病人,1例TKI治疗耐药的多发肿瘤病例尸体解剖样本。

    研究三:一位患Ⅳ期肺腺癌伴癌性胸腔积液,纵隔、肝、骨、中枢神经系统多发转移的病人。

    干预措施:

    研究一:将该家系中的Ⅲ-1和 Ⅲ-2的肿瘤标本进行EGFR基因序列分析。

    研究二:将16例病人的肿瘤样本进行RFLP分析,并对新发现的EGFR D761Y突变进行功能分析。

    研究三:对病人的肿瘤样本进行DNA序列分析,发现EGFR 19外显子缺失突变,并且是对吉非替尼治疗的敏感性突变,2004年1月起给予6个周期的卡铂、紫杉醇、吉非替尼(250 mg/d)。并通过腰椎穿刺抽取脑脊液,取得癌细胞,建立该病人的DFCILU-011细胞系。2004年6月评价疗效,发现出现多发性脑转移,而全身其他部位的病灶得到控制。病人继续予吉非替尼单药治疗并接受40 Gy的全脑放疗。尔后按照病情变化以及脑脊液中吉非替尼浓度变化逐步加大吉非替尼剂量至1 250 mg/d。病人死亡后取其肠、肺、肝、软脑膜病灶进行基因测序及DHPLC分析。

    评价指标: EGFR T790M突变分析。

    6 主要结果

    研究一: DNA测序发现Ⅲ-1、Ⅲ-2样本均有T790M杂合性突变,同时Ⅲ-3和Ⅲ-4外周血单核细胞也发现相同的突变。同时将Ⅲ-1的5个独立原发灶和Ⅲ-2的转移病灶的石蜡包埋切片提取RNA,并通过RT-PCR获得cDNA进行序列分析,结果如表1。         

    研究二:

    (1)16个TKI治疗耐药病人中有8个存在肿瘤细胞EGFR基因二次突变,其中7个为T790M突变,1个为在脑转移病灶中新发现的D761Y突变。

    (2)D761Y突变是EGFR 761位的酪氨酸被天冬氨酸替代,而该病人的外周血细胞中未发现该突变。

    (3)D761Y突变的生化及生理学性质:D761Y及L858R都存在时EGFR激酶活性与只有L858R存在时无差异。D761Y及L858R都存在较只有L858R存在时吉非替尼对293T细胞系生长的抑制能力有所下降。构建只存在L858R、同时存在D761Y及L858R以及同时存在T790M及L858R的Ba/F3细胞系,通过不同浓度的吉非替尼对该细胞系抑制效果比较,同时存在T790M及L858R较其他两组更易发生耐药。同时存在D761Y及L858R较只存在L858R时,吉非替尼抑制细胞生长效果更差。

    (4)对一个TKI治疗耐药的多发肿瘤病例尸体解剖样本进行DNA序列分析,肺、纵隔、肝、肾上腺病灶均可见A750P突变合并T790M突变,而脑病灶仅见A750P突变。

    研究三:

    (1)取该病人肿瘤样本进行DNA序列分析,结果为EGFR 19外显子缺失突变,与吉非替尼治疗敏感性相关。

    (2)取该病人胸腔积液建立DFCILU-011细胞系,吉非替尼对该细胞系的半抑制浓度(50% inhibiting concentration,IC50)为10~50 nmol/L。

    (3)主要影像学检查结果:2004年6月发现多发性脑转移但全身其他病灶均得到控制;2004年9月发现颅内进展性转移合并软脑膜侵犯但其他病灶无明显变化;2004年11月颅内病灶有所缩小但软脑膜侵犯无变化;2004年12月颅内仅存轻微的软脑膜侵犯。

    (4)在治疗过程中,增大吉非替尼的剂量,脑脊液中吉非替尼的浓度如表2。

    (5)病人因NSCLC进展于2005年3月死亡。

    (6)取病人尸体解剖肠、肺、肝、软脑膜转移病灶样本,进行EGFR 18-21外显子DHPLC及基因测序分析。肠、肺、肝样本均可发现T790M突变,而在未予吉非替尼治疗的肿瘤样本和中枢神经系统肿瘤样本中均未发现该突变。

    7 结 论

    研究一:该肺泡细胞癌家系的遗传易感性可能与EGFR T790M耐药突变有关,见图1。

    研究二: T790M突变常见于获得性TKI治疗耐药的病人,TKI耐药突变的类型和性质与肿瘤的解剖位置和TKI抑制的靶点有关。

    研究三: 增加给药剂量可提高吉非替尼在脑脊液中的浓度,达到治疗NSCLC脑转移的目的。给予吉非替尼治疗后,病人肠、肝、肺等肿瘤病灶出现T790M突变而在中枢神经系统中未见该突变的原因可能是T790M突变在中枢神经系统病灶中缺乏长期存在优势,见图2。

    8 评 论

    肺癌的治疗已从化疗时代转向分子靶向治疗时代,攻击肺癌细胞的靶点涉及到多个方面,包括:酪氨酸激酶抑制剂(TKI)、新生血管抑制剂、维甲酸受体(RXR)抑制剂、蛋白酶小体抑制剂、环氧化酶2(Cox 2)抑制剂、叶酸抑制剂等。在 NSCLC患者中,有高达40%~80%的比例发生EGFR过量表现(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患者还伴有人表皮生长因子受体-2(Her2)的过表达。

    在所有的分子靶向治疗剂中,酪氨酸激酶抑制剂倍受关注,其作用靶点是酪氨酸蛋白激酶(PTK)。TKI抑制细胞膜表面EGFR细胞内众多酪氨酸激酶的自磷酸化作用,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。TKI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,前者主要用于NSCLC的治疗,后者主要用于慢性粒细胞性白血病和胃肠间质瘤。

    目前针对信号传导分子-蛋白激酶而设计的EGFR-TKI主要有三种:吉非替尼(Gefitinib,商品名Iressa)、埃罗替尼(Erlotinib,商品名Tarceva)和拉帕替尼(Lapatinib,为EGFR和Her2双重抑制剂)。虽然NSCLC高表达EGFR,但是临床试验显示吉非替尼治疗非小细胞肺癌在西方人群只有10%左右的患者有反应,而东方人群接受吉非替尼治疗的临床疗效却高达20%~30%。为何吉非替尼的疗效比理论预期值明显偏低,并且出现种族差异?推究其因,后来发现原来吉非替尼只对带有L858R氨基酸改变或L747-A750缺失这两种突变形式造成的EGFR蛋白过量表现有明显的抑制效果,这种体细胞突变最常发生于腺癌、非吸烟者、亚洲人和女性。于是,研究非小细胞肺癌患者身上携带的EGFR突变是否异常便成为临床医师决定采用吉非替尼进行化学治疗的指征。

    为何突变的受体对TKI更敏感?正常情况下,EGFR与EGF或其他配体(如TGF琢)结合后,被激活形成二聚体,活化内源性酪氨酸激酶,EGFR胞内酪氨酸残基发生自磷酸化,EGFR激活后可激活许多的下游信号途径,如PI3K-Akt、JAK-STAT、raf-MEK-erk/MAPK等途径(见图3)。而在EGFR突变状态下,配体依赖的raf-MEK-erk/MAPK途径受阻,非配体依赖的PI3K-Akt途径起主要作用并呈现持续活化状态,作用于胞内段激酶的药物(如吉非替尼)就能对仍处于高活性状态的PI3K-Akt发挥抑制作用。此外,泛素(ubiquitin,Ubi)介导的受体降解在正常情况下是配体依赖的,在突变状况下则是非受体依赖的;而且依赖于分子伴侣Hsp90(图中红色十字架)的受体成熟受到PI3K-Akt信号的调控。因此,突变的EGFR对TKI更敏感。

    然而,科学界才替TKI药物吉非替尼找到新的出路,却又遭遇到癌细胞的反扑。Shih等[1]首先报道在EGFR-L858R突变所致药物敏感的NSCLC腺癌病人中获得了T790M突变,正是该突变导致了病人对吉非替尼的耐药。接着Bell等[2]报道一个具有NSCLC的遗传家族,6个肿瘤患者中的4个发现T790M突变,显示药物耐受性的遗传特征。EGFR-T790M突变导致吉非替尼的耐药随后被进一步的人群研究所证实[3,4]。针对这一新的情况,研究人员进一步探究发现,不同于L858R或L747-A750使得EGFR成为Gefitinib的作用目标,T790M的点突变却使EGFR能躲过Gefitinib的攻击,因此带有EGFR在T790M位置突变的癌细胞反而能突破Gefitinib的重围而不断生长[5,6],见图4。

    正所谓“成也突变,败也突变”,新的人类与疾病的斗争总是波浪式的前进。EGFR-T790M点突变的发现,一方面为我们预测药物疗效增添了新的Biomarker,另一方面新的突变或许能成为抗癌新药开发的潜在靶标。

个人公众号:treeofhope

举报 使用道具

回复 支持 0 反对 1
願願净  初中二年级 发表于 2012-12-27 12:59:12 | 显示全部楼层 来自: 浙江杭州
极大关注!
phpinfo  大学二年级 发表于 2012-12-27 21:31:35 | 显示全部楼层 来自: 北京
老马 请继续 期待。。。

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