Adoptive cell therapy for the treatment of patients with metastatic melanoma

Adoptive cell therapy for the treatment of patients with metastatic melanoma
Adoptive cell therapy for the treatment of patients with metastatic melanoma

Available online at https://www.360docs.net/doc/d6988013.html,

Adoptive cell therapy for the treatment of patients with metastatic melanoma

Steven A Rosenberg and Mark E Dudley

Adoptive cell therapy(ACT)is the best available treatment for

patients with metastatic melanoma.In a recent series of three

consecutive clinical trials using increasing lymphodepletion

before infusion of autologous tumor in?ltrating lymphocytes

(TIL),objective response rates between49%and72%were

seen.Persistence of infused cells in the circulation at one

month was highly correlated with anti-tumor response as was

the mean telomere length of the cells infused and the number of

CD8+CD27+cells infused.Responses occur at all sites and

appear to be durable with many patients in ongoing response

beyond three years.In the most recent trial of25patients

receiving maximum lymphodepletion,seven of the25patients

(28%)achieved a complete response.Of the12patients in the

three trials who achieved a complete response all but one are

ongoing between18and75months.We recently

demonstrated that ACT using autologous lymphocytes

genetically modi?ed to express anti-tumor T cell receptors can

mediate tumor regression and this approach is now being

applied to patients with common epithelial cancers.

Address

Surgery Branch,National Cancer Institute,National Institutes of Health,

Bethesda,MD,United States

Corresponding author:Rosenberg,Steven A(sar@https://www.360docs.net/doc/d6988013.html,)

Current Opinion in Immunology2009,21:233–240

This review comes from a themed issue on

Tumour Immunology

Edited by Ton Schumacher and Nick Restifo

Available online21st March2009

0952-7915/$–see front matter

Published by Elsevier Ltd.

DOI10.1016/j.coi.2009.03.002

Introduction

Melanoma appears to be unique among human cancers

because of its ability to induce signi?cant numbers of

lymphocytes with anti-tumor activity during the natural

course of tumor growth[1].Thus,tumor in?ltrating

lymphocytes(TIL)or peripheral lymphocytes repeatedly

stimulated in vitro with autologous melanoma cells often

demonstrate in vitro recognition of melanoma cells based

on assays of lysis or cytokine secretion.The ability to

isolate and characterize anti-tumor lymphocytes from

patients with melanoma has thus enabled the identi?-

cation and characterization of multiple melanoma associ-

ated antigens that can be the target of immunotherapy

[2,3].The development of techniques to grow large

numbers of anti-tumor lymphocytes has enabled the

development of cell transfer therapies that are by far,

the most effective treatment for patients with metastatic

melanoma[4,5 ,6].

Effective cancer immunotherapy is dependent on the

presence of large numbers of anti-tumor lymphocytes

with appropriate homing and effector functions that

enable them to seek out and destroy cancer cells in vivo.

Adoptive cell therapy(ACT)refers to an immunotherapy

approach in which anti-tumor lymphocytes are identi?ed

and grown ex vivo and then infused into the cancer

patient,often along with vaccines or growth factors that

can augment the in vivo impact of the transferred cells[6].

Since ACT involves the administration of ex vivo cultured

lymphocytes,very large numbers of anti-tumor lympho-

cytes can be generated and infused.The ability to test the

activity of cells before infusion enables the identi?cation

of highly selected cells with high avidity for tumor anti-

gens.Naturally occurring anti-tumor lymphocytes are

often anergized or tolerized in vivo and thus the ability

to culture these cells ex vivo away from suppressive

in?uences that exist in vivo enables the administration

of highly activated cells that exhibit multiple anti-tumor

effector functions.Perhaps the most important factor

responsible for the effectiveness of ACT is the ability

to manipulate the host before cell transfer to provide an

improved environment for the transferred cells.A variety

of immunosuppressive in?uences can exist in the cancer

patient including the presence of lymphocytes or myeloid

cells with immunosuppressive activity.These suppres-

sive factors may play a role in limiting the anti-tumor

effectiveness of non-speci?c immune stimulants such as

interleukin-2(IL-2).Cancer vaccines are ineffective

probably owing partly to the presence of these suppres-

sive in?uences[7].This ability to remove lymphoid and

myeloid suppressor cells before immune stimulation is

unique to ACT as a form of immunotherapy.

It should be emphasized that although melanoma is

unique in its ability to naturally result in the generation

of anti-tumor T cells in vivo,there does not appear to be

any difference in the susceptibility of different cancer

types to the anti-tumor activity of lymphocytes.

Virtually all tumors are equally susceptible to lysis

and are able to stimulate cytokine release from anti-

tumor lymphocytes when tumor antigen is encountered.

Thus principles that are being learned concerning the

effectiveness of ACT in patients with melanoma can be

of value in applying ACT therapy to patients with other

cancer types using lymphocytes genetically engineered to manifest anti-tumor function[6,8 ].

Early clinical studies of ACT for melanoma

A crucial step in the development of effective ACT for the treatment of patients with melanoma was the demonstration in1987that lymphocytes in?ltrating into metastatic melanoma deposits could be grown in inter-leukin-2(IL-2)and exhibit major histocompatability complex(MHC)restricted recognition of both fresh and cultured melanoma cells[1].Utilizing current tech-niques reactivity against melanoma can be detected in TIL from approximately80%of melanoma patients [9,10].The?rst use of TIL to treat patients with metastatic melanoma was reported in1988and increased numbers of patients were reported in1994 [11,12].Eighty-six patients were treated with autolo-gous TIL plus high dose IL-2.Twenty-eight patients received cells without a preparative regimen and58 patients received a preinfusion dose of25mg/kg cyclo-phosphamide.IL-2was given at720,000IU/kg every eight hours to tolerance[13].Over80%of patients received>1011cells.Objective response rates in patients with metastatic melanoma treated with cells alone or with this low dose cyclophosphamide were31% and35%respectively.Objective responses were seen in 34%of patients who had not received prior IL-2and in 32%of patients who were refractory to prior treatment with IL-2.In these initial trials single cell suspensions were generated from tumors using enzymatic digestion and grown in IL-2until suf?cient cell numbers were generated.Cells were administered irrespective of their anti-tumor activity as measured by in vitro assays although analysis of cell activity demonstrated a signi?-cant correlation between clinical response and the in vitro lysis of autologous tumor by the administered TIL [14](p=0.0008).Objective clinical responses were also associated with cells that were cultured for shorter periods of time and that had shorter doubling times (p=0.0001and0.03respectively)[15].A subgroup of patients received cells that were labeled with indium-111and traf?c of the administered cells to tumor depos-its also correlated with clinical response(p=0.022)[16–18].Although objective anti-tumor responses were seen in these early trials many of the responses were short-lived.Of the29patients who experienced an objective clinical response only?ve were complete and the median duration of all partial responders was only four months.Analysis of the survival of the transferred cells in vivo using cells genetically labeled with neomycin phosphotransferase demonstrated a lack of persistence of the infused cells[19].Less than0.1%of the cells in the circulation at one week following infusion were the transferred cells.These early studies,however,demon-strated the potential value of ACT and modi?cations of this approach have led to dramatic improvement in its effectiveness[4,5 ,6].ACT following lymphodepletion in patients with melanoma

Early animal models predicted that the effectiveness of cell transfer therapies could be improved by administering either total body irradiation or lymphodepleting che-motherapy before the cell transfer.The limited persistence of the transferred cells in our early human trials thus led us to explore the use of lymphodepletion in patients with metastatic melanoma before receiving ACT with TIL.A series of clinical trials have been performed in a total of93 patients with metastatic melanoma exploring the use of increasing levels of lymphodepletion[4,5 ,6,20].In the conduct of these recent trials a change was made in the procedures used to generate TIL for cell transfer.In the earlier trials entire excised tumors were subjected to enzymatic digestion to form a single cell suspension that was then cultured in6000IU/ml IL-2.Lymphocytes in?l-trating into the tumor stroma grew and after two to three weeks,cultures were generally cleared of tumor cells and lymphocytes were continued in culture until the target number of cells was obtained.In the earlier trials these entire populations of TIL were administered without further selection.In the current trial a modi?ed procedure was utilized[10].Excised tumors were minced into tiny fragments and individual fragments placed in the wells of a 24well culture plate,or alternatively limited numbers of cells from a single cell suspension were put in individual wells of a24well plate.All wells then were individually grown and separately tested for their ability to recognize either the autologous tumor or tumors sharing MHC anti-gens.Individual cultures showing appropriate reactivity were then further expanded,generally to a total of1010to 1011cells before they were infused into patients.Although more labor intensive,this latter technique had the advantage of identifying cultures with anti-tumor activity but potentially had the disadvantage of limiting the hetero-geneity or polyclonality of the cells administered[21,22]. Utilizing this new method of cell preparation,three consecutive protocols were performed using increasing levels of lymphodepletion(Figure1).In the?rst protocol, 43patients were treated with autologous lymphocytes following the administration of a non-myeloablative che-motherapy regimen consisting of60mg/kg cyclophospha-mide given on two consecutive days followed by?ve days of25mg/m2?udarabine.A second trial was then con-ducted in25patients in which the same chemotherapy was given(but condensed to a?ve day period)followed by200cGy whole body irradiation the day before cell administration.In a third trial in25patients the total body irradiation was intensi?ed by giving200cGy twice a day for three consecutive days for a total of1200cGy.In the latter two trials,circulating CD34+hematopoietic stem cells were administered.In all protocols720,000IU/kg IL-2was administered to tolerance.The objective response rates by RECIST criteria in the three sequential protocols were49%,52%and72%respectively(Table1).

234Tumour Immunology

Responses were often durable and many patients have ongoing responses beyond three years,and in the earliest trial beyond ?ve years.All but one of the 12complete responses are ongoing from 18to 75months.Objective cancer regressions were seen at all sites in the body including lung,liver,brain,lymph nodes,subcutaneous tissues and bone.Examples of these responses are shown in Figure 2.The overall survival of patients in these three trials is shown in Figure 3,which also includes the survival of the original 86patients who received T cell transfer without major lymphodepletion.It should be emphasized that these clinical trials are consecutive rather than randomized and thus,although it appears that increasing lymphodepletion has led to improvement in survival,this conclusion must be drawn with caution.These studies have shown that T cell based immunother-apy is capable of mediating the regression of large vascu-larized invasive metastatic melanoma in humans.It should be emphasized that the widely held belief that immunotherapy can only affect minimal disease in the adjuvant setting is thus not correct.

Mechanisms of action of ACT in humans

Extensive studies of the mechanisms of action of ACT in humans have been performed.On the basis of animal models it appears that the lymphopenic environment acts predominantly by eliminating T regulatory cells and by eliminating competition for homeostatic cytokines such as IL-7and IL-15that are vital for T cell survival [23,24,25 ].Other factors may be involved as well such as the impact of chemotherapy and whole body irradiation on increasing extravasation of bacteria from the intestinal tract and thus result in toll like receptor stimulation [26 ].Evidence for a role of IL-15in our ACT clinical trials comes from studies demonstrating that there are no circulating levels of IL-15detectable in the serum of patients before lymphodepleting chemoradiation but high levels exist in all patients at the time of cell infusion following the chemoradiation [5 ].

Extensive studies have been performed to identify both patient and cell factors that are associated with objective clinical responses.Persistence of the infused cells was highly associated with objective response (p >0.001)[22,27].In contrast to prior studies in which the persist-ence of the administered T cells was very limited,many patients in the current trials exhibited high levels of persistence,sometimes reaching up to 75%of all of the circulating CD8+cells.It was unusual for patients to exhibit a response unless there was some persistence of the administered cells although persistence alone was not a suf?cient criterion to ensure responsiveness.The median percent of the administered cells in PBMC at one to two months was 18.5%in responders compared with only 1%in non-responders.

Studies in the Pmel mouse model predicted that the state of differentiation of the transferred cells was inversely related to the effectiveness of these cells in ACT [28,29].Our studies have suggested that this is the case in humans as well.Two cell characteristics that appear to correlate with the ability of cells to mediate an anti-tumor response are long telomere length (p <0.01)and cells that highly express CD27(p <0.0001)[30,31].Both are markers of less differentiated cells.The response rate was 11%when mean telomere length was less than 5kb that contrasted

Adoptive cell therapy for the treatment of patients with metastatic melanoma Rosenberg and Dudley 235

Figure

1

Schema of the lymphodepleting preparative regimens used in the adoptive cell transfer protocols in the Surgery Branch,NCI.

Table 1

Cell transfer therapy.a Treatment Total PR

CR

OR (%)No TBI 4317(77+,45+,34+,29,28,14,13,11,8,8,7,4,3,3,2,2,2)3(75+,70+,60+,59+)21(49%)200cGy TBI 2511(45+.41+.35+.1410,6,5,5,4,3,3)2(49+,38+)

13(52%)1200cGy TBI

25

11(26+,19+,19+,19+,13,7,6,6,5,4,3)

7(29+,19,25+,25+,19+,19+,18+)

18(72%)

52responding patients:42had prior IL-2,21had prior IL-2+chemotherapy.a

All patients with metastatic melanoma received a preparative regimen of cyclophosphamide (60mg/kg/day ?2d)and ?udarabine (25mg/m 2/day ?5d)either with no total body irradiation (TBI)or with 200or 1200cGy TBI followed by the administration of autologous TIL plus IL-2(720,000IU/kg q 8h).

with a 58%response rate in patients who received cells with mean telomere greater than 5kb.

Improvements in ACT that are being studied

The 49–72%objective response rates in patients with metastatic melanoma indicate that ACT represents the best available treatment for patients with this disease [4,5 ,6].A summary of Surgery Branch,NCI immunother-apy efforts for the treatment of patients with metastatic melanoma is shown in Figure 4.Objective response rates to treatment vaccines are about 3%,IL-2and anti CTLA-4about 15%,and ACT has increased response rates to 72%with increasing lymphodepletion before cell transfer.Efforts to both improve ACT treatment as well as simplify it to make more widely available are underway.

Simpli?ed techniques for generating very ‘‘young’’TIL have been developed [32].The use of low dose out-patient IL-2(125,000IU/kg qd)[33]is also being explored as a method to potentially reduce some of the cytokine related toxicities seen with the use of high dose IL-2.

Several approaches that may enhance the effectiveness of ACT in humans are summarized in Table 2.Perhaps the single most important factor yet to be explored is the addition of vaccines to the treatment regimen.In animal models the administration of a vaccine encoding the antigen recognized by the transferred cells substan-tially improves the anti-tumor ef?cacy of treatment [34].This represents a high priority for testing in the human.

236Tumour Immunology

Figure

2

Objective clinical regressions in patients with metastatic melanoma treated with cell transfer therapy.(a)Regression of melanoma metastases in the heart (upper),adrenal (middle)and peritoneal cavity (lower)now ongoing at 34months in a 53-year-old male.(b)Regression of multiple liver

metastases now ongoing at 60months in a 45-year-old male.(c)Rapid regression of multiple subcutaneous and nodal metastases now ongoing at 35months in a 29-year-old male.(d)Regression of a large fungating scalp mass now ongoing at 34months in a 40-year-old male.

Some patients with metastatic melanoma do not have easily resectable lesions that can give rise to TIL or have lesions with too low a number of in?ltrating lymphocytes to provide TIL for therapy.These considerations led us to perform trials exploring the genetic modi?cation of autologous lymphocytes with genes encoding anti-tumor T cell receptors [6].In an initial clinical trial,circulating lymphocytes from patients with melanoma were trans-duced with retroviruses encoding an anti-MART-1T cell receptor [8 ,35].These lymphocytes were administered to patients following the cyclophosphamide/?udarabine non-myeloablative chemotherapy.In our initial report two of 17patients responded to this therapy both of whom were disease free three years later.Retroviruses have now been constructed encoding T cell receptors with higher avidity for recognition of melanoma antigens and those are now being evaluated in a clinical trial [36].T cell receptors that recognize antigenic epitopes from p53,gp100,carcinoembryonic antigen and the cancer-testes antigen,NY-ESO-1are being clinically tested as well (Figure 4)[37,38,39 ].In addition,chimeric antigen receptors have been constructed utilizing the variable region of the heavy and light chains of monoclonal antibodies against Her2/neu and against CD19(Figure 5).These chimeric receptors utilize signaling chains from CD28,41BB and CD3zeta and when retro-virally inserted into normal lymphocytes can confer the recognition capability of these antibodies [40].The ability

Adoptive cell therapy for the treatment of patients with metastatic melanoma Rosenberg and Dudley 237

Figure

3

Survival of patients treated with cell transfer therapy in four consecutive clinical trials using increasing regimens of a lymphodepleting preparative regimen before adoptive cell transfer (NMA,non-myeloablative chemotherapy;TBI,total body irradiation).The number in parentheses is the number of patients in each trial.

Figure

4

Objective response rates using RECIST criteria in patients with

metastatic melanoma treated in the Surgery Branch,NCI using different therapeutic strategies.Overall response rates in patients treated with vaccines is about 3%and with IL-2or anti-CTLA4is about 15%.With increasing levels of lymphodepletion,adding total body irradiation (TBI)and nonmyeloablative chemotherapy (NMA)to adoptive cell transfer (ACT)can achieve response rates as high as 72%.

238Tumour Immunology

Table2

Opportunities for improving ACT for the treatment of human cancer.a

Approach Selected examples

1.Genetic modi?cation of lymphocytes to introduce new recognition speci?cities8-b TCR

Chimeric TCR

2.Genetic modi?cation of lymphocytes to alter function functions of T cells Costimulatory molecules(CD8,41BB)

Cytokines(IL-2,IL-12,IL-15)

Homing molecules(CD62L,CCR7)

Prevention of apoptosis(Bcl-2)

3.Modify host lymphodepletion Selective depletion of CD4+cells or T regulatory cells

4.Block inhibitory signals on reactive lymphocytes Antibodies to CTLA-4or PD-1

5.Administer vaccines to stimulate transferred cells Recombinant virus,peptides,dendritic cells

6.Administer alternative cytokines to support cell growth IL-15,IL-21,IL-12

7.Stimulate antigen presenting cells Toll-like receptor agonists

8.Generate less differentiated lymphocytes Select effector memory cells;alter growth

promoting cytokines in vitro

9.Overcome antigen escape variants NK cells

a Modi?ed from reference[6].

Figure5

Schematic diagrams of the structure of retroviruses encoding conventional T cell receptors and chimeric T cell receptors prepared for use in adoptive cell transfer trials.

to convert normal lymphocytes into cells with anti-tumor activity has the potential to extend ACT to patients with common cancers and these clinical trials have begun. References and recommended reading

of special interest

of outstanding interest

1.Muul LM,Spiess PJ,Director EP,Rosenberg SA:Identi?cation of

speci?c cytolytic immune responses against autologous

tumor in humans bearing malignant melanoma.J Immunol

1987,138:989-995.

2.Van der Bruggen P et al.:A gene encoding an antigen

recognized by cytolytic T lymphocytes on a human melanoma.

Science1991,254:1643-1647.

3.Rosenberg SA:A new era for cancer immunotherapy based on

the genes that encode cancer antigens.Immunity1999,

10:281-287.

4.Dudley ME et al.:Cancer regression and autoimmunity in

patients after clonal repopulation with anti-tumor

lymphocytes.Science2002,298:850-854.

5. Dudley ME et al.:Adoptive cell therapy for patients with metastatic melanoma:evaluation of intensive myeloablative chemoradiation preparative regimens.J Clin Oncol2008, 26:5233-5239.

Three lymphodepleting regimens were combined with tumor reactive TIL administration and compared for safety and ef?cacy in sequential ACT clinical trials.Overall52of93patients(56%)experienced an objective response.

6.Rosenberg SA,Restifo NP,Yang JC,Morgan RA,Dudley ME:

Adoptive cell transfer:a clinical path to effective cancer

immunotherapy.Nat Rev Cancer2008,8:299-308.

7.Rosenberg SA,Yang JC,Restifo NP:Cancer immunotherapy:

moving beyond current vaccines.Nat Med2004,

10:909-915.

8. Morgan RA et al.:Cancer regression in patients after transfer of genetically engineered lymphocytes.Science2006,

314:126-129.

This is the?rst study to show that patients can be successfully treated with lymphocytes engineered by retroviral transduction to express a new TCR conferring recognition of a tumor antigen.Tumor reactive lympho-cytes were shown to persist after transfer by PCR and tetramer FACS analysis,and two patients responded to the cell and gene therapy.

9.Topalian SL,Muul LM,Rosenberg SA:Growth and immunologic

characteristics of lymphocytes in?ltrating human tumor.Surg Forum1986,37:390-391.

10.Dudley ME,Wunderlich JR,Shelton TE,Even J,Rosenberg SA:

Generation of tumor-in?ltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients.

J Immunother2003,26:332-342.

11.Rosenberg SA et al.:Use of tumor in?ltrating lymphocytes and

interleukin-2in the immunotherapy of patients with metastatic melanoma.Preliminary report.N Engl J Med1988,

319:1676-1680.

12.Rosenberg SA et al.:Treatment of patients with metastatic

melanoma using autologous tumor-in?ltrating lymphocytes

and interleukin-2.J Natl Cancer Inst1994,86:1159-1166.

13.Rosenberg SA et al.:Treatment of283consecutive patients

with metastatic melanoma or renal cell cancer using

high-dose bolus interleukin-2.JAMA1994,271:907-913.

14.Aebersold P et al.:Lysis of autologous melanoma cells by

tumor in?ltrating lymphocytes:association with clinical

response.J Natl Cancer Inst1991,13:932-937.

15.Schwartzentruber DJ et al.:In vitro predictors of therapeutic

response in melanoma patients receiving tumor in?ltrating

lymphocytes and interleukin-2.J Clin Oncol1994,

12:1475-1483.16.Fisher B et al.:Tumor localization of adoptively transferred

Indium-111labeled tumor in?ltrating lymphocytes in patients with metastatic melanoma.J Clin Oncol1989,7:250-261.

17.Grif?th KD et al.:In vivo distribution of adoptively transferred

indium-111labeled tumor in?ltrating lymphocytes and

peripheral blood lymphocytes in patients with metastatic

melanoma.J Natl Cancer Inst1989,81:1709-1717.

18.Pockaj BA et al.:Localization of Indium-111-labelled tumor

in?ltrating lymphocytes to tumor in patients receiving adoptive immunotherapy:augmentation with cyclophosphamide in

association with response.Cancer1994,73:1731-1737.

19.Rosenberg SA et al.:Gene transfer into humans:

immunotherapy of patients with advanced melanoma,using

tumor-in?ltrating lymphocytes modi?ed by retroviral gene

transduction.N Engl J Med1990,323:570-578.

20.Dudley ME et al.:Adoptive cell transfer therapy following non-

myeloablative but lymphodepleting chemotherapy for the

treatment of patients with refractory metastatic melanoma.

J Clin Oncol2005,23:2346-2357.

21.Zhou J,Dudley ME,Rosenberg SA,Robbins PF:Selective

growth,in vitro and in vitro,of individual T cell clones from

tumor-in?ltrating lymphocytes obtained from patients with

melanoma.J Immunol2004,173:7622-7629.

22.Zhou J,Dudley ME,Rosenberg SA,Robbins PF:Persistence of

multiple tumor-speci?c T cell clones is associated with

complete tumor regression in a melanoma patient receiving

adoptive cell transfer therapy.J Immunother2005,28:53-62. 23.Antony PA et al.:CD8+T cell immunity against a tumor/self-

antigen is augmented by CD4+T helper cells and hindered by naturally occurring T regulatory cells.J Immunol2005,

174:2591-2601.

24.Gattinoni L et al.:Removal of homeostatic cytokine sinks by

lymphodepletion enhances the ef?cacy of adoptively

transferred tumor-speci?c CD8+T cells.J Exp Med2005,

202:907-912.

25.

Wrzesiniski C et al.:Hematopoietic stem cells promote the

expansion and function of adoptively transferred antitumor

CD8T cells.J Clin Invest2007,117:492-501.

Genetic and immunological techniques were used in a transgenic Pmel mouse model to uncover an unexpected mechanism for the promotion of ACT effectiveness by intensive ablative regimens.Introduced hemato-poietic stem cells improved ACT ef?cacy by aiding CD8+cell engraftment and persistence.

26.

Paulos CM et al.:Microbial translocation augments the

function of adoptively transferred self/tumor-speci?c CD8+T cells via TLR4signaling.J Clin Invest2007,117:2197-2204.

A mechanisms for the improvement of ACT by radiation induced lym-phodepletion was explored in the Pmel mouse model.Antigen presenting cells activated by translocated gut micro?ora stimulated transferred tumor reactive CD8+cells,suggesting methods for improving clinical ACT therapy.

27.Robbins PF et al.:Cutting edge:persistence of transferred

lymphocyte clonotypes correlates with cancer regression in

patients receiving cell transfer therapy.J Immunol2004,

173:7125-7130.

28.Klebanoff CA et al.:Central memory self/tumor-reactive CD8+T

cells confer superior antitumor immunity compared with

effector memory T cells.Proc Natl Acad Sci USA2005,

102:9571-9576.

29.Gattinoni L et al.:Acquisition of full effector function in vitro

paradoxically impairs the in vivo antitumor ef?cacy of

adoptively transferred CD8+T cells.J Clin Invest2005,

115:1616-1626.

30.Zhou J,Shen X,Hodes RJ,Rosenberg SA,Robbins P:Telomere

length of transferred lymphocytes correlates with in vivo

persistence and tumor regression in melanoma patients

receiving cell transfer therapy.J Immunol2005,175:7046-7052.

31.Huang J et al.:Modulation by IL-2of CD70and CD27

expression on CD8+T cells:importance for the therapeutic

effectiveness of cell transfer immunotherapy.J Immunol2006, 176:7726-7735.

Adoptive cell therapy for the treatment of patients with metastatic melanoma Rosenberg and Dudley239

32.Tran KQ et al.:Minimally cultured tumor-in?ltrating

lymphocytes display optimal characteristics for adoptive cell therapy.J Immunother2008,31:742-751.

33.Yang JC et al.:Randomized study of high-dose and low-dose

interleukin-2in patients with metastatic renal cancer.

J Clin Oncol2003,21:3127-3132.

34.Overwijk WW et al.:Tumor regression and autoimmunity after

reversal of a functionally tolerant state of self-reactive CD8+T cells.J Exp Med2003,198:569-580.

35.Hughes MS et al.:Transfer of a TCR gene derived from a patient

with a marked antitumor response conveys highly active T-cell effector functions.Hum Gene Ther2005,16:457-472.

36.Johnson LA et al.:Gene transfer of tumor-reactive TCR confers

both high avidity and tumor reactivity to nonreactive

peripheral blood mononuclear cells and tumor-in?ltrating.

J Immunol2006,177:6548-6559.

37.Theoret MR et al.:Relationship of p53overexpression

on cancers and recognition by anti-p53T cell

receptor-transduced T cells.Hum Gene Ther2008,

19:1219-1231.

38.Parkhurst MR,Joo J,Riley JP,Yu Z,Li Y,Robbins PF,

Rosenberg SA:Characterization of genetically modi?ed T cell receptors that recognize the CEA:691–699peptide in the

context of HLA-A2.1on human colorectal cancer cells.

Clin Cancer Res2009,15(1):169-180.

39.

Robbins PF et al.:Single and dual amino acid substitutions in TCR CDRs can enhance antigen-speci?c T cell functions.

J Immunol2008,180:6116-6131.

Site directed mutagenesis was used to engineer modi?cations in pre-sumptive MHC-contact positions and antigen-contact positions of alpha and beta TCR genes.Increased avidity and antigen-speci?c T cell functionality was observed when modi?ed TCRs were transduced into lymphocytes.

40.Eshhar Z,Waks T,Gross G,Schindler DG:Speci?c activation

and targeting of cytotoxic lymphocytes through chimeric

single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell

receptors.Proc Natl Acad Sci USA1993,90(2):720-724.

240Tumour Immunology

工程塑料重点

第一章绪论 1 工程塑料的定义与分类 定义:能承受一定的外力作用,有良好的机械性能和尺寸稳定性,在高、低温下能保持其优异性能,可作为工程结构制件使用的塑料,包括通用工程塑料和特种工程塑料(高性能工程塑料或超级工程塑料)。 2 工程塑料的主要性能特点是什么? ⑴相对密度小⑵比强度高⑶耐热性好⑷化学稳定性好⑸电绝缘性能优良⑹机械性能优良⑺耗能少⑻尺寸稳定性好 3 工程塑料的发展方向是什么? ⑴应用创新技术改造传统工艺;⑵工程塑料的合金化和复合化是开发高性能合成材料的重要途径。⑶?零垃圾?、?零排放?、?高的资源再生率?以及?环境友好产品的生产?是21世纪规范企业环保行为的共同要求。 4 ⑴高分子材料又称聚合物材料,是以聚合物为基体组分的材料,主要包括塑料、橡胶、纤维、聚合物基复合材料、粘合剂、涂料和功能高分子等。 ⑵塑料以树脂(或在加工过程中用单体直接聚合)为主要成分,以增塑剂、填充剂、润滑剂、着色剂等添加剂为辅助成分,在加工过程中能流动成型的材料。 ⑶合成树脂是人们采用化学方法,人工合成出来的一种与天然树脂类似的有机高分子材料。 ⑷通用塑料一般指产量大、用途广、成型性好、价格便宜、力学性能一般,主要作为非结构材料使用的塑料 ⑸特种塑料一般指具有特种功能,可用于航空航天等特殊应用领域的塑料 ⑹通用工程塑料⑺特种工程塑料 ⑻热塑性塑料在特定温度范围内能反复加热软化和冷却硬化的塑料 ⑼热固性塑料在受热或其它条件下能固化或具有不溶不熔特性的塑料 5 主要的工程塑料助剂有哪些?并了解其特点。 主要工程塑料助剂包括阻燃剂、抗氧剂、光稳定剂、抗静电剂、化学发泡剂、润滑剂、偶联剂、抗电弧剂和抗电弧径迹剂、相容剂(增容剂)、成核剂。 6 谈谈你对工程塑料的合金化和复合化是开发高性能合成材料的重要途径的认识。 第二章聚酰胺 1 聚酰胺的定义、分类和命名 定义:大分子主链中含有重复结构单元酰胺基团的聚合物的统称。 分类:脂肪族聚酰胺半芳香族聚酰胺全芳香族聚酰胺含杂环芳香族聚酰胺脂环族聚酰胺 命名:⑴由内酰胺开环聚合的尼龙⑵由二元胺和二元酸缩聚得到的聚合物⑶用重复的二胺或二酸的简称表示⑷共聚尼龙是用上述方法命名的尼龙名称组合的,主要成分的尼龙名称放在前面 2 不同品种聚酰胺的各自突出性能特点分别是什么? 《1》尼龙⑴优良的力学性能⑵自润滑性、耐摩擦性好⑶优良的耐热性⑷优异的电绝缘性能⑸优良的耐候性⑹吸水性大 3 叙述缩聚型与加成型脂肪族聚酰胺分子结构的特点。两者各用什么方法制备?不同类型聚酰胺的共性所在。 4 解释透明聚酰胺具有透明性的原因。 5 说明Nomex的结构特点,解释它何以具有高强度、高耐热性。 6 说明全对位聚芳酰胺有哪两种制备路线?写出两种路线所得产物的结构,进而说明全对位聚芳酰胺结构特点和性能特点。 7 聚酰胺的化学结构与性能间关系涉及哪些方面内容?并加以分析。 8 为什么PA6和PA66具有较高的熔点? PA6和PA66都是线形大分子,化学结构规整性比较高,分子主链上没有支链,因此可以结晶;酰氨基极

(完整版)高聚物合成第一章绪论

第一章绪论 1.1 高分子材料科学的历史回顾 高分子的概念始于20世纪20年代,但应用更早。 ?1839年,美国人Goodyear发明硫化橡胶。 ?1855年,英国人Parks实现硝化纤维产业化 ?1870年海厄特用硝化纤维素与樟脑混合制得赛璐珞celluloid。 ?1884年法国人De Chardonnet(查唐纳脱)把硝化纤维素放在酒精和乙醚中得到溶液,得到人造丝。 ?1907年贝克兰发明酚醛树脂-Bakelite 第一个真正意义上人工合成的树脂是酚醛树脂,由此,打开了合成高分子的新时代。 ?1920年,德国人Staudinger发表了“论聚合”的论文,提出了高分子的概念,并预测了聚氯乙烯和聚甲基丙烯酸甲酯等聚合物的结构。 ?1935年,Carothes发明尼龙66,1938年工业化。 ?30年代,一系列烯烃类加聚物被合成出来并工业化,PVC(1927~1937),PV Ac(1936),PMMA(1927~1931),PS(1934~1937),LDPE(1939)。自由基聚合发展。 ?高分子溶液理论在30年代建立,并成功测定了聚合物的分子量。Flory为此获得诺贝尔奖。 ?40年代,二次大战促进了高分子材料的发展,一大批重要的橡胶和塑料被合成出来。丁苯橡胶(1937),丁腈橡胶(1937),丁基橡胶(1940),有机氟材料(1943),ABS(1947),涤纶树脂(1940~1950)。 ?50年代,Ziegler和Natta发明配位聚合催化剂,制得高密度PE和有规PP,低级烯烃得到利用。 ?1956年,美国人Szwarc发明活性阴离子聚合,开创了高分子结构设计的先河。 ?50年后期至60年代,大量高分子工程材料问世。聚甲醛(1956),聚碳酸酯(1957),聚砜(1965),聚苯醚(1964),聚酰亚胺(1962) ?60年代以后,特种高分子和功能高分子得到发展。 特种高分子:高强度、耐高温、耐辐射、高频绝缘、半导体等。 功能高分子:分离材料(离子交换树脂、分离膜 等)、导电高分子、感光高分子、高分子催化剂、高吸水性树脂、医用高分子、药用高分子、高分子液晶等。 ?80年代以后,新的聚合方法和新结构的聚合物不断出现和发展。 ?新的聚合方法:阳离子活性聚合、基团转移聚合、活性自由基聚合、等离子聚合等等; ?新结构的聚合物:新型嵌段共聚物、新型接枝共聚物、星状聚合物、树枝状聚合物、超支化聚合物、含C60聚合物等等。 1.2 合成高分子材料的特点 a. 原材丰富、易加工、成本低、效益高; 一座年产万吨的合成纤维厂相当于30万亩棉田或250万头绵羊的棉毛产量。 10000吨天然橡胶: 需要热带70万亩土地,载3000万株橡胶树,需5万劳力,6~8年时间; 10000吨合成橡胶: 工厂:150人。 b. 材料性能多样且优异 1.耐烧蚀; 2.吸水性; 3.疏水性; 4.阻尼性能; 5.生物性能; 6.轻质高强; 7.透明性; 8.加工性;9. 耐蚀、绝缘性

第一章 绪论

第一章绪论 第一节石油化学工业发展概况 石油是我们现代生活中必不可少的燃料和化工原料,早在三四千年前,人们就已经发现和开始利用石油了。从19世纪中叶起,近代石油工业迅速兴起,石油作为一种重要的能源、优质的有机化工原料,在世界政治经济中的地位日趋重要,在各国的国民经济和国防建设中起着举足轻重的作用。 一、石油化学工业概貌 以石油、石油炼制后的产品、油田气或天然气为原料,采取不同工艺生产燃料油、润滑油、化工原料、化工中间体和化工产品的工业,称为石油化学工业。 按加工与用途划分,石油加工业有两大分支:一是石油经过炼制生产各种燃料油、润滑油、石蜡、沥青、焦炭等石油产品;二是把石油分离成原料馏分,进行热裂解,得到基本有机原料,用于合成生产各种石油化学制品。前一分支是石油炼制工业体系,后一分支是石油化工体系。因此,通常把以石油、天然气为基础的有机合成工业,即石油和天然气为起始原料的有机化学工业称为石油化学工业,简称石油化工。炼油和化工二者是相互依存相互联系的,是一个庞大而复杂的工业部门,其产品有数千种之多。它们的相互结合和渗透,不但推动了石油化工的技术发展,也是提高石油经济效益的主要途径。石油化工是20世纪60年代以来快速发展起来的一个新兴工业部门。石油化学工业产品概貌及其石油化工上中下游产品关系如图1一1和图1一2所示。 石油化工包括以下四大生产过程:基本有机化工生产过程、有机化工生产过程、高分子化工生产过程和精细化工生产过程。基本有机化工生产过程是以石油和天然气为起始原料,经过炼制加工制得三烯(乙烯、丙烯、丁烯)、三苯(苯、甲苯、二甲苯)、乙炔和萘等基本有机原料。有机化工生产过程是在“三烯、三苯、乙炔、萘”的基础上,通过各种合成步骤制得醇、醛、酮、酸、酯、醚、腈类等有机原料。高分子化工生产过程是在有机原料的基础上,经过各种聚合、缩合步骤制得合成纤维、合成树脂、合成橡胶(即三大合成材料)等最终产品。 石油化工是精细化工的基础,精细化工的原料大部分来自廉价的石油化工。精细化工为石油化工提供高档末端材料,如催化剂、表面活性剂、油品添加剂、三大合成材料用助剂等。精细化工生产多项工业和尖端技术所需要的工程材料和功能性材料,取得高附加值。所以,一般认为精细化程度已成为衡量石化工业水平的尺度。 二、石油化学工业的历史与现状 石油化学工业是在煤化学工业的基础上发展起来的,基本有机化工原料的生产大致经过三个阶段,即初级阶段、煤化学阶段、石油化学阶段。最早,人们是以农副产品的“发酵”和“干馏”的方法获得品种有限的有机原料,如粮食发酵制取酒精,木材干馏取得甲醇、丙酮、醋酸、苯酚等。19世纪后半期,钢铁工业的发展带动了炼焦工业的发展。用煤炼焦时,副产约3%的煤焦油,煤焦油富含苯、甲苯、萘等有用芳烃,将这些芳烃提取出来,为染料生产提供了充足原料。随后,人们用焦炭和石灰石融炼出电石(CaC2 ),电石与水反应轻而易举地制得乙炔,利用乙炔的特有活性可制得氯乙烯、醋酸乙烯、氯丁二烯、三氯乙烯、丙烯腈、乙醛、异戊二烯等有机原料,再由此衍生最终产品,这是煤化工最灿烂的历史阶段。由于煤化工的工艺路线和效益不佳,在1970年以石油为燃料的化工产品比例猛增到80%以上。到1979年,在西欧天然液体及气体燃料在化工原料的能源消耗已高达99%,固体燃料只占1%。从石油化学工业的诞生开始,它就以其低成本、高产出、高效益的绝对优势,结束了仅以煤和农产品为化学工业原料的历史。 (一)世界石油石油化学工业的历史与现状 石油化学工业的兴起始于美国。C·Ellis(西·埃力斯)于1908年创建了世界上最早的石油

相关文档
最新文档